Language selection

Search

Patent 3029209 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3029209
(54) English Title: CD3 BINDING ANTIBODIES
(54) French Title: ANTICORPS SE LIANT A CD3
Status: Examination Requested
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 39/395 (2006.01)
  • C07K 16/46 (2006.01)
  • C12N 15/13 (2006.01)
(72) Inventors :
  • TRINKLEIN, NATHAN (United States of America)
  • VAN SCHOOTEN, WIM (United States of America)
  • ALDRED, SHELLEY FORCE (United States of America)
  • HARRIS, KATHERINE (United States of America)
  • PHAM, DUY (United States of America)
(73) Owners :
  • TENEOBIO, INC. (United States of America)
(71) Applicants :
  • TENEOBIO, INC. (United States of America)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2017-06-20
(87) Open to Public Inspection: 2017-12-28
Examination requested: 2022-06-20
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2017/038373
(87) International Publication Number: WO2017/223111
(85) National Entry: 2018-12-21

(30) Application Priority Data:
Application No. Country/Territory Date
62/352,698 United States of America 2016-06-21
62/394,360 United States of America 2016-09-14
62/491,908 United States of America 2017-04-28

Abstracts

English Abstract

The present invention relates to novel human CD3 antigen-binding polypeptides and their preparation and use in the treatment and/or diagnosis of various diseases, and also relates to bispecific antibody molecules capable of activating immune effector cells and their use in diagnosis and/or treatment of various diseases.


French Abstract

La présente invention concerne de nouveaux polypeptides de liaison d'antigène CD3, leurs préparation et leur utilisation dans le traitement et/ou le diagnostic de différentes maladies, et concerne en outre des molécules de type anticorps bispécifiques capables d'activer des cellules effectrices immunitaires et leur utilisation dans le diagnostic et/ou le traitement de dverses maladies.

Claims

Note: Claims are shown in the official language in which they were submitted.


What is Claimed is:
1. An antigen-binding protein specific for CD3, comprising:
a variable heavy chain domain comprising CDR1, CDR2 and CDR3 sequences in a
human VH framework, where the CDR1 sequence has a formula:
G1 G2 S3 I4 X5 S6 X7 X8 X9 x10
where X5, X7, X8, X9 and X10 may be any amino acid;
the CDR2 sequence has the formula:
I1, X2 X3, S4', G5' X6' X7,
where X2'; X3'; X8' and X7' may be any amino acid;
the CDR3 sequence has the formula
X1" R2" W3" R4" H5" D6" I7" X8" X9" X10" Y11" P12" Y13" Y14" Y15" Y16" G17"
M18" D19" V20"
where X1"; X8"; X9" and X10'' may be any amino acid;
and a variable light chain domain.
2. An antigen-binding protein specific for CD3, comprising a variable heavy
chain
domain comprising CDR1, CDR2 and CDR3 sequences in a human VH framework, where
the
CDR1, CDR2 or CDR3 are found of any of SEQ ID NO:1-68;
and a variable light chain domain.
3. An antigen-binding protein specific for CD3, comprising a variable heavy
chain
domain comprising CDR1, CDR2 and CDR3 sequences in a human VH framework
wherein the
CDR sequences are a sequence with at least 85% identity to a CDR sequence or
set of CDR
sequences in any one of SEQ ID NO:1-68;
and a variable light chain domain.
4. An antigen-binding protein specific for CD3, comprising a variable heavy
chain
domain comprising CDR1, CDR2 and CDR3 sequences in a human VH framework
wherein the
CDR sequences are a sequence with up to 3 amino acid substitutions relative to
a CDR
sequence or set of CDR sequences in any one of SEQ ID NO:1-68;
and a variable light chain domain.
42


5. The antigen-binding protein of any of claims 1-4, wherein the variable
light chain
domain comprises a CDR1, CDR2 and CDR3 sequences in a human VL framework
wherein the
CDR sequences are a sequence with up to 3 amino acid substitutions relative to
a CDR
sequence or set of CDR sequences in SEQ ID NO:69; or wherein the CDR sequences
are a
sequence with at least 85% identity to a CDR sequence or set of CDR sequences
in SEQ ID
NO:69.
6. The antigen-binding protein of any of claims 1-5, wherein the variable
heavy
chain domain comprises an amino acid sequence set forth in any of SEQ ID NO:1-
68.
7. The antigen-binding protein of any of claims 1-6, wherein the variable
light chain
domain comprises an amino acid sequence of SEQ ID NO:69.
8. An antigen-binding protein of any of claims 1-7, wherein when contacted
with T
cells in an activation assays the antigen binding protein induces release of
reduced levels of
one or both of IL-2 and I L--10 relative to a reference anti-CD3 antibody.
9. The antigen-binding protein of any of claims 1-8, further comprising an
Fc region.
10. The antigen-binding protein of claim 9, wherein the Fc region has been
engineered to reduce effector functions.
11. The antigen-binding protein of any of claims 1-10, wherein the affinity
(KD) of the
protein for human CD3 delta epsilon is around about 10 -6 M to around about 10
-11 M.
12. The antigen-binding protein of any of claims 1-10, wherein the protein
cross-
reacts with human and Cynomolgus macaque CD3 protein.
13. The antigen-binding protein of any of claims 1-12, wherein the protein
is a single
chain.
14. The antigen-binding protein of any of claims 1-12, wherein the protein
is two
chains or a multiple thereof

43


15. The antigen-binding protein of any of claims 1-12, wherein the protein
is three
chains.
16. The antigen-binding protein of any of claims 1-12, wherein the protein is
three chains
and both antigen-binding arms comprise of antibody heavy and light chains
17. The antigen-binding protein of any of claims 1-12, wherein the protein
further
comprises a variable heavy chain domain specific for a protein other than CD3.
18. The antigen-binding protein of any of claims 1-17, wherein the protein
further
comprises a variable heavy chain domain specific for a protein other than CD3;
wherein when contacted with T cells in an activation assays the antigen
binding protein
induces release of reduced levels of one or both of IL-2 and IL-10 relative to
a reference anti-
CD3 antibody; and
induces more than 30% tumor cytotoxicity in standard in vitro assays using
tumor cells
and human T cells.
19. The antigen-binding protein of claim 17, wherein the variable heavy
chain domain
specific for a protein other than CD3 is a heavy chain only domain.
20. The antigen-binding protein of claim 19, wherein the variable heavy
chain domain
specific for a protein other than CD3 further comprises a light chain variable
region.
21. The antigen-binding protein of claim 19, wherein the light chain
variable region is
the same as light chain variable region of the CD3-binding region.
22. The antigen-binding protein of any of claims 17-21, wherein the protein
other
than CD3 is a tumor associated antigen.
23. The antigen-binding protein of any of claims 17-21, wherein the protein
other
than CD3 is a pathogen antigen.
24. The antigen-binding protein of any of claims 17-21, wherein the protein
other
than CD3 is an immunoregulatory protein.

44

25. A pharmaceutical composition comprising an antigen-binding protein of
any of
claims 1-24.
26. The pharmaceutical formulation of claim 25, in a unit dose formula.
27. A polynucleotide encoding an antigen-binding protein of any of claims 1-
24.
28. A vector comprising a polynucleotide of claim 27.
29. A cell comprising a vector of claim 28.
30. A method of producing an antigen-binding protein of any of claims 1-24,

comprising growing a cell according to claim 29 under conditions permissive
for expression of
the protein, and isolating the protein from the cells.
31. A method of treatment, comprising administering to an individual an
effective
dose of a protein of an antigen-binding protein of any of claims 1-24.
32. The method of claim 31, wherein the individual is human.
33. A method of making an antigen-binding protein specific for CD3,
comprising:
immunizing OmniFlic or UniRat animals with CD3; and
identifying antigen-specific heavy chain sequences.
34. A method of making an antigen-binding protein of claim 17, comprising:
immunizing OmniFlic or UniRat animals with an antigen other than CD3; and
identifying antigen-
specific heavy chain sequences that can be combined in a molecule containing
the antigen-
binding protein of any of claims 1-16.
35. A bispecific antigen-binding protein, wherein the protein comprises of a
binding
domain specific for CD3 and a binding domain specific for an antigen other
than CD3;
wherein when contacted with T cells in an activation assay the bispecific
antigen binding
protein induces release of reduced levels of one or both of IL-2 and IL-10
relative to a reference
anti-CD3 antibody; and

induces more than 30% tumor cytotoxicity in standard in vitro assays using
tumor cells
and human T cells.
36. A bispecific antibody comprising:
(i) a CD3-binding variable region comprising CDR1, CDR2 and CDR3 sequences of
SEQ ID NO:1 in a human VH framework;
(ii) a variable light chain domain comprising CDR1, CDR2 and CDR3 sequences of
SEQ
ID NO:69 in a human VL framework;
(iii) a single chain anti-BCMA variable region comprising CDR1, CDR2 and CDR3
sequences at positions 26-33; 51-58; 97-108, respectively, of SEQ ID NO:70 in
single or in
tandem configuration.
37. The bispecific antibody of claim 36, further comprising an Fc region.
38. The bispecific antibody of claim 37, wherein the CD3-binding variable
region
comprises SEQ ID NO:1, the light chain variable region comprises SEQ ID NO:69
and the
BCMA-binding variable region comprises SEQ ID NO:70 or SEQ ID NO:71.
46

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
CD3 BINDING ANTIBODIES
CROSS REFERENCE
[0001] This application claims benefit of U.S. Provisional Patent
Application No. 62/352,698,
filed June 21, 2016, U.S. Provisional Patent Application No. 62/394,360, filed
September 14,
2016 and U.S. Provisional Patent Application No. 62/491,908, filed April 28,
2017, which
applications are incorporated herein by reference in their entirety.
BACKGROUND
[0002] The body's immune system serves as a defense against infection,
injury and cancer.
Two separate but interrelated systems, humoral and cellular immune systems,
work together to
protect the body. The humoral system is mediated by soluble factors, named
antibodies, which
neutralize products recognized as being foreign by the body. In contrast, the
cellular system
involves cells, such as T cells and macrophages, which remove and neutralize
foreign invaders.
[0003] The activation of T cells is critical for the stimulation of immune
responses. T cells exhibit
immunological specificity and direct most of the cellular immune responses.
Although T cells do
not secrete antibodies, they are required for the secretion of antibodies by B
lymphocytes. T cell
activation requires the participation of a number of cell surface molecules,
such as the T cell
receptor complex, and CD4 or CD8 molecules. The antigen-specific T cell
receptor (TcR) is
composed of a disulfide-linked heterodimer, membrane glycoprotein with chains,
alpha and beta
(a and 13), or gamma and delta (y and 6). The TcR is non-covalently linked
with a complex of
invariant proteins, designated CD3.
[0004] T cells are known to exert potent antitumor effects in numerous
experimental settings.
Antibodies capable of effectively recruiting T cells against tumor cells have
been available as
bispecific antibodies, for example directed to tumor-associated antigens
(TAAs) and agonistic T-
cell membrane proteins, such as the TCR/CD3 complex and 0D28. These bispecific
antibodies
are capable of activating T cells, irrespective of their TCR specificity,
resulting in specific lysis of
cells carrying the respective TAAs.
[0005] However, while anti-CD3 bispecific antibodies can redirect T-cell-
mediated lysis toward
malignant cells, clinical trials with CD3-based bsAbs have shown high toxicity
in patients. Non-
specific T-cell activation from bsAbs can occur in an antigen-independent
manner due to the
Fc/Fc receptor (FcR) interaction, or in an antigen-dependent manner when
antigen is expressed
on both normal and tumor cells. Both mechanisms may have been responsible for
the toxicity
observed in prior clinical studies. (See for example, Link et al. (1998) Int.
J. Cancer 77(2):251-6;
1

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
Durben et al. Molecular Therapy (2015); 23 4, 648-655). Because of the
resulting cytokine
release syndrome, there have been significant blocks to the development of
these antibodies for
therapeutic purposes.
[0006] For example, Mack et al. (1995) PNAS 92:7021-7025 reported a
bispecific single chain
(bssc)-antibody with CD3-specificity, designated a bispecific T-cell engager
(BiTE). However, as
in earlier studies with different anti-CD3 containing bispecific antibodies,
excessive T-cell
activation and cytokine release induced upon in vivo application limits the
safely applicable
doses to less than 100 pg/day resulting in serum concentrations below 1 ng/ml.
[0007] CD3 specific antibodies, and bispecific antibodies derived therefrom
are provided by the
invention.
Publications
[0008] CD3 antibodies are disclosed, for example, in U.S. Pat. Nos.
5,585,097; 5,929,212;
5,968,509; 6,706,265; 6,750,325; 7,381,803; 7,728,114. Bispecific antibodies
with CD3 binding
specificity are disclosed, for example, in U.S. Pat. Nos. 7,262,276;
7,635,472; 7,862,813; and
8,236,308, each herein specifically incorporated by reference.
SUMMARY
[0009] Compositions and methods of use thereof are provided for a family of
closely related
antibodies that bind to and activate signaling through CD3, e.g. activation of
CD3 + T cells. The
antibodies within the family comprise a set of CDR sequences as defined
herein. The family of
antibodies provides a number of benefits that contribute to utility as
clinically therapeutic
agent(s). The antibodies within the family include members with a range of
binding affinities,
allowing the selection of a specific sequence with a desired affinity. The
ability to fine tune
affinity is of particular importance to manage the level of CD3 activation in
an individual being
treated, and thereby reduce toxicity. Members of the antibody family may have
an affinity (KD)
for CD3 ranging from around about 10-6 to around about 10-11 M. Certain
members of the
antibody family of the invention are cross-reactive with CD3 protein of
Cynomolgus macaque,
and a specific motif required for this cross-reactivity is identified,
allowing the selection of
antibodies for pre-clinical or clinical testing on this basis.
[0010] Anti-CD3 antibodies that have affinities (KD) of 50 nM or greater,
100 nM or greater, 500
nM or greater, or 1 .M or greater can be desirable to more closely mimic the
TCR/MHC
interaction and minimize toxic cytokine release while maintaining effective
tumor cell lysis. In
some embodiments, anti-CD3 antibodies are characterized or selected for
reduced propensity
2

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
to induce cytokine release, upon binding to a competent T cell, e.g. for
release of IL-2 and IFNy.
Antibodies may be selected for therapeutic use that optimize killing of tumor
cells and reduced
release of cytokines, e.g. an antibody that, within the family of antibody
sequences described
herein, induces a cytokine release that is less than about half the maximum
observed for a
family member in a comparative assay, and may be less, e.g. less and about 25%
the maximum
observed for a family member in a comparative assay.
[0011] In some embodiments, bispecific or multispecific antibodies are
provided, which
comprise at least a heavy chain variable region from the antibody family of
the invention and
may comprise a heavy and light chain variable region provided herein.
Bispecific antibodies
comprise at least the heavy chain variable region of an antibody specific for
a protein other than
CD3, and may comprise a heavy and light chain variable region. In some such
embodiments,
the second antibody specificity binds to a tumor associated antigen, a
targeting antigen, e.g.
integrins, etc., a pathogen antigen, a checkpoint protein, and the like.
Various formats of
bispecific antibodies are within the ambit of the invention, including without
limitation single
chain polypeptides, two chain polypeptides, three chain polypeptides, four
chain polypeptides,
and multiples thereof.
[0012] The family of CD3 specific antibodies of the invention comprise a VH
domain,
comprising CDR1, CDR2 and CDR3 sequences in a human VH framework. The CDR
sequences may be situated, as an example, in the region of around amino acid
residues 26-35;
53-59; and 98-117 for CDR1, CDR2 and CDR3, respectively, of the provided
exemplary variable
region sequences set forth in SEQ ID NO:1-68. It will be understood by one of
skill in the art
that the CDR sequences may be in different position if a different framework
sequence is
selected, although generally the order of the sequences will remain the same.
[0013] The CDR sequences for an antibody of the invention may have the
following sequence
formulas. An X indicates a variable amino acid, which may be specific amino
acids as indicated
below.
CDR1
G1 G2 S3 14 X5 S6 X7 X8 X9 X10
where:
X6 may be any amino acid; in some embodiments X6 is S or R;
X7 and X8 may be any amino acid; in some embodiments X7 and X8 are,
independently,
S or G. In some embodiments X7X8 are SS, or GG;
X9 may be any amino acid; in some embodiments X9 is H or Y; in some
embodiments X9
is H;
3

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
X10 may be any amino acid, in some embodiments X10 is Y or F; in some
embodiments
X10 is Y.
In some embodiments, the CDR1 sequence has the formula: GG s I x5 SHFIG Y,
where X5 is as defined above. In some embodiments a CDR1 sequence of an anti-
CD3
antibody of the invention comprises the sequence set forth in any of SEQ ID
NO:1-68, residues
26-35.
CDR2
X3, S4, Gs, X6r X7r
where:
X2, may be any amino acid; in some embodiments X2, is S, Y or H;
X3, may be any amino acid; in some embodiments X3, is Y, H or R;
X6, may be any amino acid; in some embodiments X6, is S, N or I or R;
X7, may be any amino acid; in some embodiments X7, is T or P.
In some embodiments the CDR2 sequence has the formula: I x2, x3, S G S T; or
I X2, x3, s GNP where X2, and X3, are as defined above. In some embodiments a
CDR2
sequence of an anti-CD3 antibody of the invention comprises the sequence set
forth in any of
SEQ ID NO:1-68, residues 53-59.
CDR3
X1" R2" W.3" R4" Hs" D6" 17" X8" X9" X10" Y11" P12" Y13" Y14" Y15" Y16" G17"
M18" D19" V20"
where:
X1,, may be any amino acid; in some embodiments X1,, is A or G.
X8,, may be any amino acid; in some embodiments X8,, is L or F.
X9,, may be any amino acid; in some embodiments X9,, is T or A
Xvy, may be any amino acid; in some embodiments Xvy, is G, A or R.
In some embodiments X.5,,X9 Xvy, are F A A, which motif corresponds to
antibodies that
cross-react with the CD3 protein of Cynomolgus macaque. In other embodiments
X8,, X9,, Xvy, are
L T A. In some embodiments a CDR3 sequence of an anti-CD3 antibody of the
invention
comprises the sequence set forth in any of SEQ ID NO:1-68, residues 98-117.
[0014] In some embodiments the CD3-binding VH domain is paired with a light
chain variable
region domain. In some such embodiments the light chain is a fixed light
chain. In some
embodiments the light chain comprises a VL domain with CDR1, CDR2 and CDR3
sequences
in a human VL framework. The CDR sequences may be those contained in SEQ ID
NO:69. In
4

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
some embodiments, the CDR sequence comprises amino acid residues 27-32; 50-52;
89-97 for
CDR1, CDR2, CDR3, respectively.
[0015] In some embodiments the CDR sequences of an antibody of the
invention are a
sequence with at least 85% identity, at least 90% identity, at least 95%
identity, at least 99%
identity relative to a CDR sequence or set of CDR sequences in any one of SEQ
ID NO:1-69. In
some embodiments a CDR sequence of the invention comprises one, two, three or
more amino
acid substitutions relative to a CDR sequence or set of CDR sequences in any
one of SEQ ID
NO: 1-69. In some embodiments said amino acid substitution(s) are one or more
of position 5
or 10 of CDR1, position 2, 6 or 7 of CDR2, position 1, 8, 9 or 10 of CDR3,
relative to the
formulas provided above.
[0016] In some embodiments, a bispecific antibody of the invention
comprises a CD3-binding
variable region described herein, paired with a light chain. In some
embodiments the light chain
comprises the variable region sequence set forth in SEQ ID NO:69, or a
variable region
comprising the set of CDR sequences in SEQ ID NO:69 and framework sequences.
Various Fc
sequences find use, including without limitation human IgG1, IgG2a, IgG2b,
IgG3, IgG4, etc. In
some embodiments, the second arm of the bispecific antibody comprises a
variable region that
specifically binds to a tumor-associated antigen. In some embodiments, the
second arm of the
bispecific antibody comprises a variable region that specifically binds to
BCMA. In some
embodiments the anti-BCMA arm is a single chain variable region, for example
as shown in
Figure 2B. In some embodiments the anti-BCMA arm comprises the variable region
sequence
set forth in SEQ ID NO:70; or the tandem variable region sequence set forth in
SEQ ID NO:71.
The Fc sequence of the anti-BCMA arm may be, without limitation, human IgG1,
IgG2a, IgG2b,
IgG3, IgG4, etc. The CDR sequences may be those contained in SEQ ID NO:70. In
some
embodiments, the CDR sequence comprises amino acid residues 26-33; 51-58; 97-
108 for
CDR1, CDR2, CDR3, respectively.
[0017] In other embodiments, pharmaceutical compositions are provided,
comprising at least a
CD3-binding VH domain of the invention, e.g. a monospecific, bispecific, etc.
antibody or
antibody-like protein comprising at least a CD3-binding VH domain of the
invention; and a
pharmaceutically acceptable excipient. The composition may be lyophilized,
suspended in
solution, etc. and may be provided in a unit dose formulation.
[0018] In some embodiments, a method is provided for treatment of cancer,
the method
comprising administering to an individual in need thereof an effective dose of
a mono-specific,
bi-specific, etc. antibody of the invention. Where the antibody is bispecific,
a second antigen-
binding site may specifically bind a tumor antigen, a checkpoint protein, etc.
In various

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
embodiments, the cancer is selected from the group consisting of ovarian
cancer, breast
cancer, gastrointestinal, brain cancer, head and neck cancer, prostate cancer,
colon cancer,
lung cancer, leukemia, lymphoma, sarcoma, carcinoma, neural cell tumors,
squamous cell
carcinomas, germ cell tumors, metastases, undifferentiated tumors, seminomas,
melanomas,
myelomas, neuroblastomas, mixed cell tumors, and neoplasias caused by
infectious agents.
[0019] In some embodiments, a method is provided for treatment of
infectious disease, the
method comprising administering to an individual in need thereof an effective
dose of a mono-
specific, bi-specific, etc. antibody of the invention. Where the antibody is
bispecific, a second
antigen-binding site may specifically bind a pathogen antigen, e.g. bacteria,
viruses or parasites.
[0020] In other embodiments, a method is provided for the production of a
bispecific antibody of
the present invention comprising expressing the antibody sequences, e.g. one
or more light
chain encoding sequences, one or more heavy chain encoding sequences, in a
single host cell.
In various embodiments, the host cell may be a prokaryotic or an eukaryotic
cell, such as a
mammalian cell.
BRIEF DESCRIPTION OF THE DRAWINGS
[0021] The invention is best understood from the following detailed
description when read in
conjunction with the accompanying drawings. The patent or application file
contains at least one
drawing executed in color. Copies of this patent or patent application
publication with color
drawing(s) will be provided by the Office upon request and payment of the
necessary fee. It is
emphasized that, according to common practice, the various features of the
drawings are not to-
scale. On the contrary, the dimensions of the various features are arbitrarily
expanded or
reduced for clarity. Included in the drawings are the following figures.
[0022] FIG. 1A-1B. (FAM1_aCD3_CDR_seqalign). FIG. 1A shows an alignment of
CDR1, 2
and 3 regions of all members of an antibody family recognizing human CD3. The
CDR
sequences correspond to amino acid residues 26-35; 53-59; and 98-117 for CDR1,
CDR2 and
CDR3, respectively, of the variable region sequences set forth in SEQ ID NO:1-
68. FIG. 1B
shows the CDR1, 2 and 3 regions of the fixed light chain (SEQ ID NO:69); and
an exemplary
anti-BCMA sequence (SEQ ID NO:70 and SEQ ID NO:71).
[0023] FIG. 2A-2E. Schematic models of bispecific human antibodies. FIG. 2A
anti-CD3:anti-
tumor-antigen bispecific antibody with common light chain (3 total unique
chains). FIG. 2B anti-
CD3:anti-tumor-antigen bispecific antibody with 2 unique light chains (4 total
unique chains).
FIG. 2C anti-CD3:anti-tumor-antigen bispecific antibody with heavy-chain only
tumor antigen
binding domain chain (3 unique chains). FIG. 20 anti-CD3:anti-tumor-antigen
bispecific antibody
6

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
with scFv tumor antigen binding domain (3 total unique chains). FIG. 2E anti-
CD3:anti-tumor-
antigen bispecific antibody with scFv anti-CD3 binding domain (3 total unique
chains).
[0024] FIG. 3. Activation of human CD8+ T cells by an a-CD3/a-PD-L1
bispecific FlicAb.
Purified human CD8+ T cells were co-cultures with a bispecific at the
indicated concentrations
and tumor cells. Ramos (B cell lymphoma) is a cell line negative for PD-L1 and
HDLM2
(Multiple Myeloma) is a cell line positive for PD-L1. 0D69 is a membrane
molecule which is
upregulated on activated T cells. Mean Fluorescent Intensity (MFI) of human
CD8+ T cells
stained with a fluorescent-labeled anti-0D69 monoclonal antibody correlates
with degree of
activation. Activation was dependent on the presence of both tumor cells
expressing PD-L1 and
bispecific.
[0025] FIG. 4. Cytolysis of tumor cells by an a-CD3/a-PD-L1 bispecific
FlicAb. Tumor cells
(HDLM2) were incubated with purified human CD8+ T cells and bispecific
antibodies. HDLM2
cells do not express CD20 and co-culture with an a-CD3/a-CD20 bispecific
FlicAb did not lead
to killing of HDLM2 cells. Only co-culture of human CD8+ T cells and HDLM2
with an a-CD3/a-
PD-L1 bispecific FlicAb lead to significant killing. HDLM2 express PD-L1 on
their surface.
[0026] FIG. 5. The table summarizes the behavior of anti-CD3 antibodies in
monospecific and
bispecific format. Column 1 shows the sequence ID for the anti-CD3 VH
sequence. Column 2
shows the MFI value for Jurkat cell binding of the parental monospecific anti-
CD3. Column 3
shows the MFI value for cyno T-cell binding of the parental monospecific anti-
CD3. Column 4
shows the name of the aCD3:aBCMA bispecific antibody. Column 5 shows the
picograms of IL-
2 released by pan T-cells stimulated by the bispecific antibody binding the
BCMA protein coated
on plastic at the dose indicated. Column 6 shows the picograms of IL-6
released by pan T-cells
stimulated by the bispecific antibody binding the BCMA protein coated on
plastic at the dose
indicated. Column 7 shows the picograms of IL-10 released by pan T-cells
stimulated by the
bispecific antibody binding the BCMA protein coated on plastic at the dose
indicated. Column 8
shows the picograms of IFN-y released by pan T-cells stimulated by the
bispecific antibody
binding the BCMA protein coated on plastic at the dose indicated. Column 9
shows the
picograms of TNFa released by pan T-cells stimulated by the bispecific
antibody binding the
BCMA protein coated on plastic at the dose indicated. Column 10 shows the EC50
of bispecific
antibody-mediated U266 tumor cell lysis in presence of human pan T-cells.
Column 11 shows
the percent lysis of U266 tumor cells in the presence of bispecific antibody
and human pan T-
cells at a dose of 333ng/mL of bispecific antibody. Column 12 shows the
protein binding affinity
of the anti-CD3 arm of the bispecific antibody measured by Octet. Column 13
shows the MFI
value for Jurkat cell binding of the bispecific antibody.
7

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
[0027] FIG. 6. Bispecific antibody-mediated tumor cell lysis.
Four aCD3_famtaBCMA
bispecific antibodies, each with a unique anti-CD3 arm and a common anti-BCMA
arm, were
tested for the ability to kill U266 BCMA+ tumor cells through redirection of
activated primary T
cells. In this experiment U266 cells that express BCMA were mixed with
activated pan T-cells in
a 10:1 E:T ratio along with the addition of bispecific antibody. The x-axis
shows the
concentration of antibody used and the y-axis shows the % lysis of tumor cells
6 hours after
addition of antibody.
[0028] FIG. 7. Bispecific U266 killing activity correlated with IL-2
release. A comparison of
bispecific antibody-mediated tumor cell lysis activity with IL-2 cytokine
release is shown in the
scatter plot. The correlation between IL-2 production and U266 tumor cell
lysis is R2= 0.37.
[0029] FIG. 8. Bispecific U266 killing activity correlated with IFN-y
release. A comparison of
bispecific antibody-mediated tumor cell lysis activity with IFN-g cytokine
release is shown in the
scatter plot. The correlation between IFN-y production and U266 tumor cell
lysis is R2= 0.53.
[0030] FIG. 9. Bispecific U266 killing activity correlated with anti-CD3
binding affinity. A
comparison of bispecific antibody-mediated U266 tumor cell lysis activity with
anti-CD3 binding
affinity is shown in the scatter plot. The correlation between U266 killing
EC50 and protein
binding affinity is R2= 0.93.
[0031] FIG. 10A-10D. Bispecific antibody-mediated tumor cell lysis.
aCD3_F1F:aBCMA
bispecific antibodies were assayed for the ability to kill three different
BCMA+ tumor cells and
one BCMA-negative cell line through redirection of activated primary T cells.
The antibodies
were comprised of an aCD3 arm (SEQ ID NO:1 and SEQ ID NO:69) and an aBCMA arm
(SEQ
ID NO:70, or SEQ ID NO:71). In this experiment, tumor cells were mixed with
activated pan T-
cells in a 10:1 E:T ratio along with the addition of bispecific antibody. FIG.
10A shows killing of
RPMI-8226 cells, FIG. 10B shows killing of NCI-H929 cells, FIG. 10C shows
killing of U-266
cells, and FIG. 100 shows killing of K562 cells, a negative control. The x-
axis shows the
concentration of antibody used and the y-axis shows the % lysis of tumor cells
6 hours after
addition of antibody.
[0032] FIG. 11A-11D. Bispecific antibody-mediated IL-2 release. The level
of IL-2 cytokine
release was measured after resting human T cells were cultured with various
tumor cell lines
and increasing doses of aCD3_F1F:aBCMA bispecific antibody (as in Figure 10).
FIG. 11A
shows IL-2 release stimulated by RPMI-8226 cells, FIG. 11B shows IL-2 release
stimulated by
NCI-H929 cells, FIG. 11C shows IL-2 release stimulated by U-266 cells, and
FIG. 1D shows IL-
2 release stimulated by K562 cells, a negative control.
8

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
[0033] FIG. 12A-120. Bispecific antibody-mediated IFN-y release. The level
of IFN-y cytokine
release was measured after resting human T cells were cultured with various
tumor cell lines
and increasing doses of aCD3_F1F:aBCMA bispecific antibody (as in Figure 10).
FIG. 12A
shows IFN-y release stimulated by RPMI-8226 cells, FIG. 12B shows IFN-y
release stimulated
by NCI-H929 cells, FIG. 1C shows IFN-g release stimulated by U-266 cells, and
FIG. 120
shows IFN-y release stimulated by K562 cells, a negative control.
DETAILED DESCRIPTION OF INVENTION
[0034] To facilitate an understanding of the invention, a number of terms
are defined below.
[0035] Before the present active agents and methods are described, it is to
be understood that
this invention is not limited to the particular methodology, products,
apparatus and factors
described, as such methods, apparatus and formulations may, of course, vary.
It is also to be
understood that the terminology used herein is for the purpose of describing
particular
embodiments only, and is not intended to limit the scope of the present
invention which will be
limited only by appended claims.
[0036] It must be noted that as used herein and in the appended claims, the
singular forms "a,"
"and," and "the" include plural referents unless the context clearly dictates
otherwise. Thus, for
example, reference to "a drug candidate" refers to one or mixtures of such
candidates, and
reference to "the method" includes reference to equivalent steps and methods
known to those
skilled in the art, and so forth.
[0037] Unless defined otherwise, all technical and scientific terms used
herein have the same
meaning as commonly understood by one of ordinary skill in the art to which
this invention
belongs. All publications mentioned herein are incorporated herein by
reference for the purpose
of describing and disclosing devices, formulations and methodologies which are
described in
the publication and which might be used in connection with the presently
described invention.
[0038] Where a range of values is provided, it is understood that each
intervening value, to the
tenth of the unit of the lower limit unless the context clearly dictates
otherwise, between the
upper and lower limit of that range and any other stated or intervening value
in that stated range
is encompassed within the invention. The upper and lower limits of these
smaller ranges may
independently be included in the smaller ranges is also encompassed within the
invention,
subject to any specifically excluded limit in the stated range. Where the
stated range includes
one or both of the limits, ranges excluding either both of those included
limits are also included
in the invention.
9

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
[0039]
In the following description, numerous specific details are set forth to
provide a more
thorough understanding of the present invention. However, it will be apparent
to one of skill in
the art that the present invention may be practiced without one or more of
these specific details.
In other instances, well-known features and procedures well known to those
skilled in the art
have not been described in order to avoid obscuring the invention.
[0040]
Generally, conventional methods of protein synthesis, recombinant cell
culture and
protein isolation, and recombinant DNA techniques within the skill of the art
are employed in the
present invention. Such techniques are explained fully in the literature, see,
e.g., Maniatis,
Fritsch & Sambrook, Molecular Cloning: A Laboratory Manual (1982); Sambrook,
Russell and
Sambrook, Molecular Cloning: A Laboratory Manual (2001); Harlow, Lane and
Harlow, Using
Antibodies: A Laboratory Manual: Portable Protocol No. I, Cold Spring Harbor
Laboratory
(1998); and Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring
Harbor Laboratory;
(1988).
Definitions
[0041] By "comprising" it is meant that the recited elements are
required in the
composition/method/kit, but other elements may be included to form the
composition/method/kit
etc. within the scope of the claim.
[0042] By "consisting essentially of", it is meant a limitation of the
scope of composition or
method described to the specified materials or steps that do not materially
affect the basic and
novel characteristic(s) of the subject invention.
[0043] By "consisting of', it is meant the exclusion from the
composition, method, or kit of any
element, step, or ingredient not specified in the claim.
[0044]
The terms "treatment", "treating" and the like are used herein to generally
mean obtaining
a desired pharmacologic and/or physiologic effect. The effect may be
prophylactic in terms of
completely or partially preventing a disease or symptom thereof and/or may be
therapeutic in
terms of a partial or complete cure for a disease and/or adverse effect
attributable to the
disease. "Treatment" as used herein covers any treatment of a disease in a
mammal, and
includes: (a) preventing the disease from occurring in a subject which may be
predisposed to
the disease but has not yet been diagnosed as having it; (b) inhibiting the
disease, i.e., arresting
its development; or (c) relieving the disease, i.e., causing regression of the
disease. The
therapeutic agent may be administered before, during or after the onset of
disease or injury.
The treatment of ongoing disease, where the treatment stabilizes or reduces
the undesirable

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
clinical symptoms of the patient, is of particular interest. Such treatment is
desirably performed
prior to complete loss of function in the affected tissues. The subject
therapy may be
administered during the symptomatic stage of the disease, and in some cases
after the
symptomatic stage of the disease.
[0045] A "therapeutically effective amount" is intended for an amount of
active agent which is
necessary to impart therapeutic benefit to a subject. For example, a
"therapeutically effective
amount" is an amount which induces, ameliorates or otherwise causes an
improvement in the
pathological symptoms, disease progression or physiological conditions
associated with a
disease or which improves resistance to a disorder.
[0046] The terms "subject," "individual," and "patient" are used
interchangeably herein to refer
to a mammal being assessed for treatment and/or being treated. In an
embodiment, the
mammal is a human. The terms "subject," "individual," and "patient" encompass,
without
limitation, individuals having cancer, individuals with autoimmune diseases,
with pathogen
infections, and the like. Subjects may be human, but also include other
mammals, particularly
those mammals useful as laboratory models for human disease, e.g. mouse, rat,
etc.
[0047] The terms "cancer," "neoplasm," and "tumor" are used interchangeably
herein to refer to
cells which exhibit autonomous, unregulated growth, such that they exhibit an
aberrant growth
phenotype characterized by a significant loss of control over cell
proliferation. Cells of interest
for detection, analysis, or treatment in the present application include
precancerous (e.g.,
benign), malignant, pre-metastatic, metastatic, and non-metastatic cells.
Cancers of virtually
every tissue are known. The phrase "cancer burden" refers to the quantum of
cancer cells or
cancer volume in a subject. Reducing cancer burden accordingly refers to
reducing the number
of cancer cells or the cancer volume in a subject. The term "cancer cell" as
used herein refers
to any cell that is a cancer cell or is derived from a cancer cell e.g. clone
of a cancer cell. Many
types of cancers are known to those of skill in the art, including solid
tumors such as
carcinomas, sarcomas, glioblastomas, melanomas, lymphomas, myelomas, etc., and
circulating
cancers such as leukemias, including specifically B cell leukemias, T cell
leukemias, etc.
Examples of cancer include but are not limited to, ovarian cancer, breast
cancer, colon cancer,
lung cancer, prostate cancer, hepatocellular cancer, gastric cancer,
pancreatic cancer, cervical
cancer, ovarian cancer, liver cancer, bladder cancer, cancer of the urinary
tract, thyroid cancer,
renal cancer, carcinoma, melanoma, head and neck cancer, and brain cancer.
[0048] "Antibody-dependent cell-mediated cytotoxicity" and "ADCC" refer to
a cell-mediated
reaction in which nonspecific cytotoxic cells that express Fc receptors, such
as natural killer
11

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
cells, neutrophils, and macrophages, recognize bound antibody on a target cell
and cause lysis
of the target cell. ADCC activity may be assessed using methods, such as those
described in
U.S. Pat. No. 5,821,337. ADCP refers to antibody dependent cell-mediated
phagocytosis.
[0049] "Effector cells" are leukocytes which express one or more constant
region receptors and
perform effector functions.
[0050] A "cytokine" is a protein released by one cell to act on another
cell as an intercellular
mediator.
[0051] "Non-immunogenic" refers to a material that does not initiate,
provoke or enhance an
immune response where the immune response includes the adaptive and/or innate
immune
responses.
[0052] The term "isolated" means that the material is removed from its
original environment
(e.g., the natural environment if it is naturally occurring). For example, a
naturally-occurring
polynucleotide or polypeptide present in a living animal is not isolated, but
the same
polynucleotide or polypeptide, separated from some or all of the coexisting
materials in the
natural system, is isolated. Such polynucleotides could be part of a vector
and/or such
polynucleotides or polypeptides could be part of a composition, and still be
isolated in that such
vector or composition is not part of its natural environment.
[0053] "Pharmaceutically acceptable excipient" means an excipient that is
useful in preparing a
pharmaceutical composition that is generally safe, non-toxic, and desirable,
and includes
excipients that are acceptable for veterinary use as well as for human
pharmaceutical use. Such
excipients can be solid, liquid, semisolid, or, in the case of an aerosol
composition, gaseous.
[0054] "Pharmaceutically acceptable salts and esters" means salts and
esters that are
pharmaceutically acceptable and have the desired pharmacological properties.
Such salts
include salts that can be formed where acidic protons present in the compounds
are capable of
reacting with inorganic or organic bases. Suitable inorganic salts include
those formed with the
alkali metals, e.g. sodium and potassium, magnesium, calcium, and aluminum.
Suitable organic
salts include those formed with organic bases such as the amine bases, e.g.,
ethanolamine,
diethanolamine, triethanolamine, tromethamine, N methylglucamine, and the
like. Such salts
also include acid addition salts formed with inorganic acids (e.g.,
hydrochloric and hydrobromic
acids) and organic acids (e.g., acetic acid, citric acid, maleic acid, and the
alkane- and arene-
sulfonic acids such as methanesulfonic acid and benzenesulfonic acid).
Pharmaceutically
acceptable esters include esters formed from carboxy, sulfonyloxy, and
phosphonoxy groups
present in the compounds, e.g., 01_6 alkyl esters. When there are two acidic
groups present, a
12

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
pharmaceutically acceptable salt or ester can be a mono-acid-mono-salt or
ester or a di-salt or
ester; and similarly where there are more than two acidic groups present, some
or all of such
groups can be salified or esterified. Compounds named in this invention can be
present in
unsalified or unesterified form, or in salified and/or esterified form, and
the naming of such
compounds is intended to include both the original (unsalified and
unesterified) compound and
its pharmaceutically acceptable salts and esters. Also, certain compounds
named in this
invention may be present in more than one stereoisomeric form, and the naming
of such
compounds is intended to include all single stereoisomers and all mixtures
(whether racemic or
otherwise) of such stereoisomers.
[0055] The terms "pharmaceutically acceptable", "physiologically tolerable"
and grammatical
variations thereof, as they refer to compositions, carriers, diluents and
reagents, are used
interchangeably and represent that the materials are capable of administration
to or upon a
human without the production of undesirable physiological effects to a degree
that would
prohibit administration of the composition.
[0056] "Homology" between two sequences is determined by sequence identity.
If two
sequences, which are to be compared with each other, differ in length,
sequence identity
preferably relates to the percentage of the nucleotide residues of the shorter
sequence which
are identical with the nucleotide residues of the longer sequence. Sequence
identity can be
determined conventionally with the use of computer programs such as the
Bestfit program
(Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer
Group,
University Research Park, 575 Science Drive Madison, Wis. 53711). Bestfit
utilizes the local
homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2
(1981), 482-
489, in order to find the segment having the highest sequence identity between
two sequences.
When using Bestfit or another sequence alignment program to determine whether
a particular
sequence has for instance 95% identity with a reference sequence of the
present invention, the
parameters are preferably so adjusted that the percentage of identity is
calculated over the
entire length of the reference sequence and that homology gaps of up to 5% of
the total number
of the nucleotides in the reference sequence are permitted. When using
Bestfit, the so-called
optional parameters are preferably left at their preset ("default") values.
The deviations
appearing in the comparison between a given sequence and the above-described
sequences of
the invention may be caused for instance by addition, deletion, substitution,
insertion or
recombination. Such a sequence comparison can preferably also be carried out
with the
program "fa5ta20u66" (version 2.0u66, September 1998 by VVilliam R. Pearson
and the
13

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
University of Virginia; see also W. R. Pearson (1990), Methods in Enzymology
183, 63-98,
appended examples and http://workbench.sdsc.edu/). For this purpose, the
"default" parameter
settings may be used.
[0057] "Variant" refers to polypeptides having amino acid sequences that
differ to some extent
from a native sequence polypeptide. Ordinarily, amino acid sequence variants
will possess at
least about 80% sequence identity, more preferably, at least about 90%
homologous by
sequence. The amino acid sequence variants may possess substitutions,
deletions, and/or
insertions at certain positions within the reference amino acid sequence.
[0058] The term "vector," as used herein, is intended to refer to a nucleic
acid molecule capable
of transporting another nucleic acid to which it has been linked. One type of
vector is a
"plasmid", which refers to a circular double stranded DNA loop into which
additional DNA
segments may be ligated. Another type of vector is a viral vector, wherein
additional DNA
segments may be ligated into the viral genome. Certain vectors are capable of
autonomous
replication in a host cell into which they are introduced (e.g., bacterial
vectors having a bacterial
origin of replication and episomal mammalian vectors). Other vectors (e.g.,
non-episomal
mammalian vectors) can be integrated into the genome of a host cell upon
introduction into the
host cell, and thereby are replicated along with the host genome. Moreover,
certain vectors are
capable of directing the expression of genes to which they are operably
linked. Such vectors are
referred to herein as "recombinant expression vectors" (or simply,
"recombinant vectors"). In
general, expression vectors of utility in recombinant DNA techniques are often
in the form of
plasmids. In the present specification, "plasmid" and "vector" may be used
interchangeably as
the plasmid is the most commonly used form of vector.
[0059] The term "host cell" (or "recombinant host cell"), as used herein,
is intended to refer to a
cell that has been genetically altered, or is capable of being genetically
altered by introduction of
an exogenous polynucleotide, such as a recombinant plasmid or vector. It
should be understood
that such terms are intended to refer not only to the particular subject cell
but to the progeny of
such a cell. Because certain modifications may occur in succeeding generations
due to either
mutation or environmental influences, such progeny may not, in fact, be
identical to the parent
cell, but are still included within the scope of the term "host cell" as used
herein.
[0060] "Binding affinity" generally refers to the strength of the sum total
of noncovalent
interactions between a single binding site of a molecule (e.g., an antibody or
other binding
molecule) and its binding partner (e.g., an antigen or receptor). The affinity
of a molecule X for
its partner Y can generally be represented by the dissociation constant (Kd).
Affinity can be
14

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
measured by common methods known in the art, including those described herein.
Low-affinity
antibodies bind antigen (or receptor) weakly and tend to dissociate readily,
whereas high-affinity
antibodies bind antigen (or receptor) more tightly and remain bound longer.
[0061] Unless specifically indicated to the contrary, the term "conjugate"
as described and
claimed herein is defined as a heterogeneous molecule formed by the covalent
attachment of
one or more antibody fragment(s) to one or more polymer molecule(s), wherein
the
heterogeneous molecule is water soluble, i.e. soluble in physiological fluids
such as blood, and
wherein the heterogeneous molecule is free of any structured aggregate. A
conjugate of
interest is PEG. In the context of the foregoing definition, the term
"structured aggregate" refers
to (1) any aggregate of molecules in aqueous solution having a spheroid or
spheroid shell
structure, such that the heterogeneous molecule is not in a micelle or other
emulsion structure,
and is not anchored to a lipid bilayer, vesicle or liposome; and (2) any
aggregate of molecules in
solid or insolubilized form, such as a chromatography bead matrix, that does
not release the
heterogeneous molecule into solution upon contact with an aqueous phase.
Accordingly, the
term "conjugate" as defined herein encompasses the aforementioned
heterogeneous molecule
in a precipitate, sediment, bioerodible matrix or other solid capable of
releasing the
heterogeneous molecule into aqueous solution upon hydration of the solid.
[0062] The word "label" when used herein refers to a detectable compound or
composition
which is conjugated directly or indirectly to the antibody. The label may
itself be detectable by
itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an
enzymatic label, may
catalyze chemical alteration of a substrate compound or composition which is
detectable.
[0063] By "solid phase" is meant a non-aqueous matrix to which the antibody
of the present
invention can adhere. Examples of solid phases encompassed herein include
those formed
partially or entirely of glass (e.g. controlled pore glass), polysaccharides
(e.g., agarose),
polyacrylamides, polystyrene, polyvinyl alcohol and silicones. In certain
embodiments,
depending on the context, the solid phase can comprise the well of an assay
plate; in others it is
a purification column (e.g. an affinity chromatography column). This term also
includes a
discontinuous solid phase of discrete particles, such as those described in
U.S. Pat. No.
4,275,149.
[0064] Antibodies, also referred to as immunoglobulins, conventionally
comprise at least one
heavy chain and one light, where the amino terminal domain of the heavy and
light chains is
variable in sequence, hence is commonly referred to as a variable region
domain, or a variable

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
heavy (VH) or variable light (VH) domain. The two domains conventionally
associate to form a
specific binding region, although as well be discussed here, specific binding
can also be
obtained with heavy chain only variable sequences, and a variety of non-
natural configurations
of antibodies are known and used in the art.
[0065] A "functional" or "biologically active" antibody or antigen-binding
molecule (including
heavy chain only antibodies and bispecific three-chain antibody-like molecules
(TCAs) herein) is
one capable of exerting one or more of its natural activities in structural,
regulatory, biochemical
or biophysical events. For example, a functional antibody or other binding
molecule, e.g. TCA,
may have the ability to specifically bind an antigen and the binding may in
turn elicit or alter a
cellular or molecular event such as signaling transduction or enzymatic
activity. A functional
antibody or other binding molecule, e.g. TCA, may also block ligand activation
of a receptor or
act as an agonist or antagonist. The capability of an antibody or other
binding molecule, e.g.
TCA, to exert one or more of its natural activities depends on several
factors, including proper
folding and assembly of the polypeptide chains.
[0066] The term "antibody" herein is used in the broadest sense and
specifically covers
monoclonal antibodies, polyclonal antibodies, monomers, dimers, multimers,
multispecific
antibodies (e.g., bispecific antibodies), heavy chain only antibodies, three
chain antibodies,
single chain Fv, nanobodies, etc., and also include antibody fragments, so
long as they exhibit
the desired biological activity (Miller et al (2003) Jour. of Immunology
170:4854-4861).
Antibodies may be murine, human, humanized, chimeric, or derived from other
species.
[0067] The term antibody may reference a full-length heavy chain, a full
length light chain, an
intact immunoglobulin molecule; or an immunologically active portion of any of
these
polypeptides, i.e., a polypeptide that comprises an antigen binding site that
immunospecifically
binds an antigen of a target of interest or part thereof, such targets
including but not limited to,
cancer cell or cells that produce autoimmune antibodies associated with an
autoimmune
disease. The immunoglobulin disclosed herein may comprise any suitable Fc
region, including
without limitation, human or other mammalian, e.g. cynomogulus, IgG, IgE, IgM,
IgD, IgA, IgG1,
IgG2, IgG3, IgG4, IgA1 and IgA2 or subclass of immunoglobulin molecule,
including engineered
subclasses with altered Fc portions that provide for reduced or enhanced
effector cell activity.
The immunoglobulins can be derived from any species. In one aspect, the
immunoglobulin is of
largely human origin.
[0068] The term "variable" refers to the fact that certain portions of the
variable domains differ
extensively in sequence among antibodies and are used in the binding and
specificity of each
particular antibody for its particular antigen. However, the variability is
not evenly distributed
16

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
throughout the variable domains of antibodies. It is concentrated in three
segments called
hypervariable regions both in the light chain and the heavy chain variable
domains. The more
highly conserved portions of variable domains are called the framework regions
(FRs). The
variable domains of native heavy and light chains each comprise four FRs,
largely adopting a
beta-sheet configuration, connected by three hypervariable regions, which form
loops
connecting, and in some cases forming part of, the beta-sheet structure. The
hypervariable
regions in each chain are held together in close proximity by the FRs and,
with the
hypervariable regions from the other chain, contribute to the formation of the
antigen-binding
site of antibodies (see Kabat et al (1991) Sequences of Proteins of
Immunological Interest, 5th
Ed. Public Health Service, National Institutes of Health, Bethesda, Md.). The
constant domains
are not involved directly in binding an antibody to an antigen, but exhibit
various effector
functions, such as participation of the antibody in antibody dependent
cellular cytotoxicity
(ADCC).
[0069] The term "hypervariable region" when used herein refers to the amino
acid residues of
an antibody which are responsible for antigen-binding. The hypervariable
region may comprise
amino acid residues from a "complementarity determining region" or "CDR",
and/or those
residues from a "hypervariable loop". "Framework Region" or "FR" residues are
those variable
domain residues other than the hypervariable region residues as herein
defined.
[0070] Variable regions of interest include at least one CDR sequence from
the variable regions
provided herein, usually at least 2 CDR sequences, and more usually 3 CDR
sequences.
exemplary CDR designations are shown herein, however one of skill in the art
will understand
that a number of definitions of the CDRs are commonly in use, including the
Kabat definition
(see "Zhao et al. A germline knowledge based computational approach for
determining antibody
complementarity determining regions." Mol lmmunol. 2010;47:694-700), which is
based on
sequence variability and is the most commonly used. The Chothia definition is
based on the
location of the structural loop regions (Chothia et al. "Conformations of
immunoglobulin
hypervariable regions." Nature. 1989;342:877-883). Alternative CDR definitions
of interest
include, without limitation, those disclosed by Honegger, "Yet another
numbering scheme for
immunoglobulin variable domains: an automatic modeling and analysis tool." J
Mol Biol.
2001;309:657-670; Ofran et al. "Automated identification of complementarity
determining
regions (CDRs) reveals peculiar characteristics of CDRs and B cell epitopes."
J lmmunol.
2008;181:6230-6235; Almagro "Identification of differences in the specificity-
determining
residues of antibodies that recognize antigens of different size: implications
for the rational
design of antibody repertoires." J Mol Recognit. 2004;17:132-143; and Padlanet
al.
17

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
"Identification of specificity-determining residues in antibodies." Faseb J.
1995;9:133-139., each
of which is herein specifically incorporated by reference.
[0071] The term "monoclonal antibody" as used herein refers to an antibody
obtained from a
population of substantially homogeneous antibodies, i.e., the individual
antibodies comprising
the population are identical except for possible naturally occurring mutations
that may be
present in minor amounts. Monoclonal antibodies are highly specific, being
directed against a
single antigenic site. Furthermore, in contrast to polyclonal antibody
preparations, which include
different antibodies directed against different determinants (epitopes), each
monoclonal
antibody is directed against a single determinant on the antigen. In addition
to their specificity,
the monoclonal antibodies are advantageous in that they may be synthesized
uncontaminated
by other antibodies. The modifier "monoclonal" indicates the character of the
antibody as being
obtained from a substantially homogeneous population of antibodies, and is not
to be construed
as requiring production of the antibody by any particular method.
[0072] The antibodies herein specifically include "chimeric" antibodies in
which a portion of the
heavy and/or light chain is identical with or homologous to corresponding
sequences in
antibodies derived from a particular species or belonging to a particular
antibody class or
subclass, while the remainder of the chain(s) is identical with or homologous
to corresponding
sequences in antibodies derived from another species or belonging to another
antibody class or
subclass, as well as fragments of such antibodies, so long as they exhibit the
desired biological
activity (U.S. Pat. No. 4,816,567; and Morrison et al (1984) Proc. Natl. Acad.
Sci. USA, 81:6851-
6855). Chimeric antibodies of interest herein include "primatized" antibodies
comprising variable
domain antigen-binding sequences derived from a non-human primate (e.g., Old
World Monkey,
Ape etc) and human constant region sequences.
[0073] An "intact antibody chain" as used herein is one comprising a full
length variable region
and a full length constant region. An intact "conventional" antibody comprises
an intact light
chain and an intact heavy chain, as well as a light chain constant domain (CL)
and heavy chain
constant domains, CH1, hinge, CH2 and CH3 for secreted IgG. Other isotypes,
such as IgM or
IgA may have different CH domains. The constant domains may be native sequence
constant
domains (e.g., human native sequence constant domains) or amino acid sequence
variants
thereof. The intact antibody may have one or more "effector functions" which
refer to those
biological activities attributable to the Fc constant region (a native
sequence Fc region or amino
acid sequence variant Fc region) of an antibody. Examples of antibody effector
functions include
C1q binding; complement dependent cytotoxicity; Fc receptor binding; antibody-
dependent cell-
mediated cytotoxicity (ADCC); phagocytosis; and down regulation of cell
surface receptors.
18

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
Constant region variants include those that alter the effector profile,
binding to Fc receptors, and
the like.
[0074] Depending on the amino acid sequence of the constant domain of their
heavy chains,
intact antibodies can be assigned to different "classes." There are five major
classes of intact
immunoglobulin antibodies: IgA, IgD, IgE, IgG, and IgM, and several of these
may be further
divided into "subclasses" (isotypes), e.g., IgG1, IgG2, IgG3, IgG4, IgA, and
IgA2. The heavy-
chain constant domains that correspond to the different classes of antibodies
are called a, 6, c,
y, and p, respectively. The subunit structures and three-dimensional
configurations of different
classes of immunoglobulins are well known. Ig forms include hinge-
modifications or hingeless
forms (Roux et al (1998) J. lmmunol. 161:4083-4090; Lund et al (2000) Eur. J.
Biochem.
267:7246-7256; US 2005/0048572; US 2004/0229310). The light chains of
antibodies from any
vertebrate species can be assigned to one of two clearly distinct types,
called K and A, based on
the amino acid sequences of their constant domains.
[0075] A "functional Fc region" possesses an "effector function" of a
native-sequence Fc region.
Exemplary effector functions include C1q binding; CDC; Fc-receptor binding;
ADCC; ADCP;
down-regulation of cell-surface receptors (e.g., B-cell receptor), etc. Such
effector functions
generally require the Fc region to be interact with a receptor, e.g. the
FcyRI; FcyRIIA; FcyRIIB1;
FcyRIIB2; FcyRIIIA; FcyRIIIB receptors, and the law affinity FcRn receptor;
and can be assessed
using various assays as disclosed, for example, in definitions herein. A
"dead" Fc is one that
has been mutagenized to retain activity with respect to, for example,
prolonging serum half-life,
but which does not activate a high affinity Fc receptor.
[0076] A "native-sequence Fc region" comprises an amino acid sequence
identical to the amino
acid sequence of an Fc region found in nature. Native-sequence human Fc
regions include a
native-sequence human IgG1 Fc region (non-A and A allotypes); native-sequence
human IgG2
Fc region; native-sequence human IgG3 Fc region; and native-sequence human
IgG4 Fc
region, as well as naturally occurring variants thereof.
[0077] A "variant Fc region" comprises an amino acid sequence that differs
from that of a
native-sequence Fc region by virtue of at least one amino acid modification,
preferably one or
more amino acid substitution(s). Preferably, the variant Fc region has at
least one amino acid
substitution compared to a native-sequence Fc region or to the Fc region of a
parent
polypeptide, e.g., from about one to about ten amino acid substitutions, and
preferably from
about one to about five amino acid substitutions in a native-sequence Fc
region or in the Fc
region of the parent polypeptide. The variant Fc region herein will preferably
possess at least
19

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
about 80% homology with a native-sequence Fc region and/or with an Fc region
of a parent
polypeptide, and most preferably at least about 90% homology therewith, more
preferably at
least about 95% homology therewith.
[0078] Variant Fc sequences may include three amino acid substitutions in
the CH2 region to
reduce FcyRI binding at EU index positions 234, 235, and 237 (see Duncan et
al., (1988) Nature
332:563). Two amino acid substitutions in the complement C1q binding site at
EU index
positions 330 and 331 reduce complement fixation (see Tao et al., J. Exp. Med.
178:661 (1993)
and Canfield and Morrison, J. Exp. Med. 173:1483 (1991)). Substitution into
human IgG1 of
IgG2 residues at positions 233-236 and IgG4 residues at positions 327, 330 and
331 greatly
reduces ADCC and CDC (see, for example, Armour KL. etal., 1999 Eur J lmmunol.
29(8):2613-
24; and Shields RL. etal., 2001. J Biol Chem. 276(9):6591-604). Other Fc
variants are possible,
including without limitation one in which a region capable of forming a
disulfide bond is deleted,
or in which certain amino acid residues are eliminated at the N-terminal end
of a native Fc form
or a methionine residue is added thereto. Thus, in one embodiment of the
invention, one or
more Fc portions of the scFc molecule can comprise one or more mutations in
the hinge region
to eliminate disulfide bonding. In yet another embodiment, the hinge region of
an Fc can be
removed entirely. In still another embodiment, the molecule can comprise an Fc
variant.
[0079] Further, an Fc variant can be constructed to remove or substantially
reduce effector
functions by substituting, deleting or adding amino acid residues to effect
complement binding
or Fc receptor binding. For example, and not limitation, a deletion may occur
in a complement-
binding site, such as a C1q-binding site. Techniques of preparing such
sequence derivatives of
the immunoglobulin Fc fragment are disclosed in International Patent
Publication Nos. WO
97/34631 and WO 96/32478. In addition, the Fc domain may be modified by
phosphorylation,
sulfation, acylation, glycosylation, methylation, farnesylation, acetylation,
amidation, and the
like.
[0080] The Fc may be in the form of having native sugar chains, increased
sugar chains
compared to a native form or decreased sugar chains compared to the native
form, or may be in
an aglycosylated or deglycosylated form. The increase, decrease, removal or
other modification
of the sugar chains may be achieved by methods common in the art, such as a
chemical
method, an enzymatic method or by expressing it in a genetically engineered
production cell
line. Such cell lines can include microorganisms, e.g. Pichia Pastoris, and
mammalians cell line,
e.g. CHO cells, that naturally express glycosylating enzymes. Further,
microorganisms or cells
can be engineered to express glycosylating enzymes, or can be rendered unable
to express
glycosylation enzymes (See e.g., Hamilton, et al., Science, 313:1441 (2006);
Kanda, et al, J.

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
Biotechnology, 130:300 (2007); Kitagawa, et al., J. Biol. Chem., 269 (27):
17872 (1994); Ujita-
Lee et al., J. Biol. Chem., 264 (23): 13848 (1989); lmai-Nishiya, et al, BMC
Biotechnology 7:84
(2007); and WO 07/055916). As one example of a cell engineered to have altered
sialylation
activity, the alpha-2,6-sialyltransferase 1 gene has been engineered into
Chinese Hamster
Ovary cells and into sf9 cells. Antibodies expressed by these engineered cells
are thus
sialylated by the exogenous gene product. A further method for obtaining Fc
molecules having a
modified amount of sugar residues compared to a plurality of native molecules
includes
separating said plurality of molecules into glycosylated and non-glycosylated
fractions, for
example, using lectin affinity chromatography (See e.g., WO 07/117505). The
presence of
particular glycosylation moieties has been shown to alter the function of
lmmunoglobulins. For
example, the removal of sugar chains from an Fc molecule results in a sharp
decrease in
binding affinity to the C1q part of the first complement component Cl and a
decrease or loss in
antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent
cytotoxicity
(CDC), thereby not inducing unnecessary immune responses in vivo. Additional
important
modifications include sialylation and fucosylation: the presence of sialic
acid in IgG has been
correlated with anti-inflammatory activity (See e.g., Kaneko, et al, Science
313:760 (2006)),
whereas removal of fucose from the IgG leads to enhanced ADCC activity (See
e.g., Shoj-
Hosaka, et al, J. Biochem., 140:777 (2006)).
[0081] In alternative embodiments, antibodies of the invention may have
an Fc sequence with
enhanced effector functions, e.g. by increasing their binding capacities to
FcyRIIIA and
increasing ADCC activity. For example, fucose attached to the N-linked glycan
at Asn-297 of Fc
sterically hinders the interaction of Fc with FcyRIIIA, and removal of fucose
by glyco-
engineering can increase the binding to FcyRIIIA, which translates into >50-
fold higher ADCC
activity compared with wild type IgG1 controls. Protein engineering, through
amino acid
mutations in the Fc portion of IgG1, has generated multiple variants that
increase the affinity of
Fc binding to FcyRIIIA. Notably, the triple alanine mutant 5298A/E333A/K334A
displays 2-fold
increase binding to FcyRIIIA and ADCC function. 5239D/I332E (2X) and
5239D/1332E/A330L
(3X) variants have a significant increase in binding affinity to FcyRIIIA and
augmentation of
ADCC capacity in vitro and in vivo. Other Fc variants identified by yeast
display also showed the
improved binding to FcyRIIIA and enhanced tumor cell killing in mouse
xenograft models. See,
for example Liu et al. (2014) JBC 289(6):3571-90, herein specifically
incorporated by reference.
[0082] The term "Fc-region-comprising antibody" refers to an antibody
that comprises an Fc
region. The C-terminal lysine (residue 447 according to the EU numbering
system) of the Fc
region may be removed, for example, during purification of the antibody or by
recombinant
21

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
engineering the nucleic acid encoding the antibody. Accordingly, an antibody
having an Fc
region according to this invention can comprise an antibody with or without
K447.
[0083] "Fv" is the minimum antibody fragment, which contains a complete
antigen-recognition
and antigen-binding site. The CD3 binding antibodies of the invention comprise
a dimer of one
heavy chain and one light chain variable domain in tight, non-covalent
association; however
additional antibodies, e.g. for use in a multi-specific configuration, may
comprise a VH in the
absence of a VL sequence. Even a single variable domain (or half of an Fv
comprising only
three hypervariable regions specific for an antigen) has the ability to
recognize and bind antigen,
although the affinity may be lower than that of two domain binding site.
[0084] The Fab fragment also contains the constant domain of the light
chain and the first
constant domain (CH1) of the heavy chain. Fab' fragments differ from Fab
fragments by the
addition of a few residues at the carboxy terminus of the heavy chain CH1
domain including one
or more cysteines from the antibody hinge region. Fab'-SH is the designation
herein for Fab' in
which the cysteine residue(s) of the constant domains bear at least one free
thiol group.
F(ab1)2antibody fragments originally were produced as pairs of Fab' fragments
which have hinge
cysteines between them. Other chemical couplings of antibody fragments are
also known.
[0085] "Humanized" forms of non-human (e.g., rodent) antibodies, including
single chain
antibodies, are chimeric antibodies (including single chain antibodies) that
contain minimal
sequence derived from non-human immunoglobulin. See, for example, Jones et al,
(1986)
Nature 321:522-525; Chothia et al (1989) Nature 342:877; Riechmann et al
(1992) J. Mol. Biol.
224, 487-499; Foote and Winter, (1992) J. Mol. Biol. 224:487-499; Presta et al
(1993) J.
lmmunol. 151, 2623-2632; Werther et al (1996) J. lmmunol. Methods 157:4986-
4995; and
Presta et al (2001) Thromb. Haemost. 85:379-389. For further details, see U.S.
Pat. Nos.
5,225,539; 6,548,640; 6,982,321; 5,585,089; 5,693,761; 6,407,213; Jones et al
(1986) Nature,
321:522-525; and Riechmann et al (1988) Nature 332:323-329.
[0086] The term "single chain antibody" as used herein means a single
polypeptide chain
containing one or more antigen binding domains that bind an epitope of an
antigen, where such
domains are derived from or have sequence identity with the variable region of
an antibody
heavy or light chain. Parts of such variable region may be encoded by VH or VL
gene segments,
D and JH gene segments, or JL gene segments. The variable region may be
encoded by
rearranged VHDJH, VLDJH, VHJL, or VLJL gene segments. V-, D- and J-gene
segments may be
22

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
derived from humans and various animals including birds, fish, sharks,
mammals, rodents, non-
human primates, camels, lamas, rabbits and the like.
[0087] The CD3-binding antibodies of the invention find particular utility
in multi-specific
configurations, which include without limitation bispecific antibodies,
trifunctional antibodies, etc.
A large variety of methods and protein configurations are known and use in
bispecific
monoclonal antibodies (BsMAB), tri-specific antibodies, etc.
[0088] First-generation BsMAbs consisted of two heavy and two light chains,
one each from two
different antibodies. The two Fab regions are directed against two antigens.
The Fc region is
made up from the two heavy chains and forms the third binding site with the Fc
receptor on
immune cells (see for example Lindhofer et al., The Journal of Immunology, Vol
155, p 219-225,
1995). The antibodies may be from the same or different species. For example,
cell lines
expressing rat and mouse antibodies secrete functional bispecific Ab because
of preferential
species-restricted heavy and light chain pairing. In other embodiments the Fc
regions are
designed to only fit together in specific ways.
[0089] Other types of bispecific antibodies include chemically linked Fabs,
consisting only of the
Fab regions. Two chemically linked Fab or Fab2 fragments form an artificial
antibody that binds
to two different antigens, making it a type of bispecific antibody. Antigen-
binding fragments (Fab
or Fab2) of two different monoclonal antibodies are produced and linked by
chemical means like
a thioether (see Glennie, M J et al., Journal of immunology 139, p 2367-75,
1987; Peter
Borchmann et al., Blood, Vol. 100, No. 9, p 3101-3107, 2002).
[0090] Various other methods for the production of multivalent artificial
antibodies have been
developed by recombinantly fusing variable domains of two antibodies. A single-
chain variable
fragment (scFv) is a fusion protein of the variable regions of the heavy (VH)
and light chains
(VL) of immunoglobulins, connected with a short linker peptide of ten to about
25 amino acids.
The linker is usually rich in glycine for flexibility, as well as serine or
threonine for solubility, and
can either connect the N-terminus of the VH with the C-terminus of the VL, or
vice versa.
Bispecific single-chain variable fragments (di-scFvs, bi-scFvs) can be
engineered by linking two
scFvs with different specificities. A single peptide chain with two VH and two
VL regions is
produced, yielding bivalent scFvs.
[0091] Bispecific tandem scFvs are also known as bi-specific T-cell
engagers (BiTEs).
Bispecific scFvs can be created with linker peptides that are too short for
the two variable
regions to fold together (about five amino acids), forcing scFvs to dimerize.
This type is known
as diabodies (Adams et al., British journal of cancer 77, p 1405-12, 1998).
The Dual-Affinity Re-
23

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
Targeting (DART) platform technology (Macrogenics, Rockville, Md.). This
fusion protein
technology uses two single-chain variable fragments (scFvs) of different
antibodies on a single
peptide chain of about 55 kilodaltons. SCORPION Therapeutics (Emergent
Biosolutions, Inc.,
Seattle, Wash.) combines two antigen-binding domains in a single chain
protein. One binding
domain is on the C-terminus and a second binding domain on the N-terminus of
an effector
domain, based on immunoglobulin Fc regions.
[0092] Tetravalent and bispecific antibody-like proteins also include DVD-
Igs which are
engineered from two monoclonal antibodies (Wu, C. et al., Nature
Biotechnology, 25, p 1290-
1297, 2007). To construct the DVD-Ig molecule, the V domains of the two mAbs
are fused in
tandem by a short linker (TVAAP) with the variable domain of the first
antibody light (VL) chain
at the N terminus, followed by the other antibodies VL and Ck to form the DVD-
Ig protein light
chain. Similarly, the variable regions of the heavy (VH) chain of the two mAbs
are fused in
tandem by a short linker (ASTKGP) with the first antibody at the N terminus,
followed by the
other antibody and the heavy chain constant domains to form the DVD-Ig protein
heavy chain
(VH1/VL1). All light chain and heavy chain constant domains are preserved in
the DVD-Ig
design, as they are critical for the formation of a disulfide-linked full IgG-
like molecule.
Cotransfection of mammalian cells with expression vectors encoding the DVD-Ig
light chain and
heavy chain leads to the secretion of a single species of an IgG-like molecule
with molecular
weight of approximately 200 kDa. This molecule has now four binding sites, 2
from each mAb.
[0093] The term "bispecific three-chain antibody like molecule" or "TCA" is
used herein to refer
to antibody-like molecules comprising, consisting essentially of, or
consisting of three
polypeptide subunits, two of which comprise, consist essentially of, or
consist of one heavy and
one light chain of a monoclonal antibody, or functional antigen-binding
fragments of such
antibody chains, comprising an antigen-binding region and at least one CH
domain. This heavy
chain/light chain pair has binding specificity for a first antigen. The third
polypeptide subunit
comprises, consists essentially of, or consists of a heavy chain only antibody
comprising an Fc
portion comprising CH2 and/or CH3 and/or CH4 domains, in the absence of a CH1
domain, and
an antigen binding domain that binds an epitope of a second antigen or a
different epitope of the
first antigen, where such binding domain is derived from or has sequence
identity with the
variable region of an antibody heavy or light chain. Parts of such variable
region may be
encoded by VH and/or VL gene segments, D and JH gene segments, or JL gene
segments. The
variable region may be encoded by rearranged VHDJH, VLDJH, VHJL, or VLJL gene
segments.
[0094] A TCA protein makes use of a heavy chain only antibody" or "heavy
chain antibody" or
"heavy chain polypeptide" as used herein means a single chain antibody
comprising heavy
24

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
chain CH2 and/or CH3 and/or CH4 but no CH1 domain. In one embodiment, the
heavy chain
antibody is composed of an antigen-binding domain, at least part of a hinge
region and CH2 and
CH3 domains. In another embodiment, the heavy chain antibody is composed of an
antigen-
binding domain, at least part of a hinge region and a CH2 domain. In a further
embodiment, the
heavy chain antibody is composed of an antigen-binding domain, at least part
of a hinge region
and a CH3 domain. Heavy chain antibodies in which the CH2 and/or CH3 domain is
truncated
are also included herein. In a further embodiment the heavy chain is composed
of an antigen
binding domain, and at least one CH (CH1, CH2, CH3, or CH4) domain but no
hinge region.
The heavy chain only antibody can be in the form of a dimer, in which two
heavy chains are
disulfide bonded other otherwise, covalently or non-covalently attached with
each other. The
heavy chain antibody may belong to the IgG subclass, but antibodies belonging
to other
subclasses, such as IgM, IgA, IgD and IgE subclass, are also included herein.
In a particular
embodiment, the heavy chain antibody is of the IgG1, IgG2, IgG3, or IgG4
subtype, in particular
I gG 1 subtype.
[0095] Heavy chain antibodies constitute about one fourth of the IgG
antibodies produced by
the camelids, e.g. camels and llamas (Hamers-Casterman C., et al. Nature. 363,
446-448
(1993)). These antibodies are formed by two heavy chains but are devoid of
light chains. As a
consequence, the variable antigen binding part is referred to as the VHH
domain and it
represents the smallest naturally occurring, intact, antigen-binding site,
being only around 120
amino acids in length (Desmyter, A., et al. J. Biol. Chem. 276, 26285-26290
(2001)). Heavy
chain antibodies with a high specificity and affinity can be generated against
a variety of
antigens through immunization (van der Linden, R. H., et al. Biochim. Biophys.
Acta. 1431, 37-
46 (1999)) and the VHH portion can be readily cloned and expressed in yeast
(Frenken, L. G.
J., et al. J. Biotechnol. 78, 11-21 (2000)). Their levels of expression,
solubility and stability are
significantly higher than those of classical F(ab) or Fv fragments (Ghahroudi,
M. A. et al. FEBS
Lett. 414, 521-526 (1997)). Sharks have also been shown to have a single VH-
like domain in
their antibodies termed VNAR. (Nuttall et al. Eur. J. Biochem. 270, 3543-3554
(2003); Nuttall et
al. Function and Bioinformatics 55, 187-197 (2004); Dooley et al., Molecular
Immunology 40,
25-33 (2003)).
[0096] An antibody or antigen-binding molecule, including the heavy chain
only antibodies and
bispecific three-chain antibody-like molecules (TCAs) herein, "which binds" an
antigen of
interest, is one that binds the antigen with sufficient affinity such that the
antibody or binding
molecule is useful as a diagnostic and/or therapeutic agent in targeting the
antigen, and does

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
not significantly cross-react with other proteins. In such embodiments, the
extent of binding of
the antibody or other binding molecule to a non-targeted antigen will be no
more than 10% as
determined by fluorescence activated cell sorting (FACS) analysis or
radioimmunoprecipitation
(RIA).
PROTEINS OF THE INVENTION
[0097] The present invention provides a family of closely related
antibodies that bind to and
activate signaling through CD3, e.g. activation of CD3 + T cells. The
antibodies within the family
comprise a set of CDR sequences as defined herein, and are exemplified by the
provided VH
sequences of SEQ ID NO:1-68, and the exemplified VL sequence of SEQ ID NO:69.
The family
of antibodies provides a number of benefits that contribute to utility as
clinically therapeutic
agent(s). The antibodies within the family include members with a range of
binding affinities,
allowing the selection of a specific sequence with a desired affinity. The
ability to fine tune
affinity is of particular importance to manage the level of CD3 activation in
an individual being
treated, and thereby reduce toxicity. For example, if low abundant tumor
antigens (less than
10,000 molecules per cell) are targeted, it is anticipated that high affinity
CD3 binders (<30nM)
are . If highly abundant tumor antigens (more than 50,000 molecules per cell)
are targeted,
CD3 binders with low affinities (>50nM) are preferred.
[0098] A suitable antibody may be selected from the library for development
and use, including
without limitation use as a bispecific antibody. Determination of affinity for
a candidate protein
can be performed using methods known in the art, e.g. Biacore measurements,
etc. Members of
the antibody family may have an affinity for CD3 with a Kd of from about 10-6
to around about
10-11, including without limitation: from about 10-6 to around about 10-10;
from about 10-6 to
around about 10-9; from about 10-6 to around about 10-8; from about 10-8 to
around about 10-11;
from about 10-8 to around about 10-10; from about 10-8 to around about 10-9;
from about 10-9 to
around about 10-11; from about i09 to around about 10-10; or any value within
these ranges. The
affinity selection may be confirmed with a biological assessment for
activation of T cells in, for
example, and in vitro or pre-clinical model, and assessment of potential
toxicity.
[0099] Certain members of the antibody family of the invention are cross-
reactive with CD3
protein of Cynomolgus macaque, and a specific motif required for this cross-
reactivity is
identified, allowing the selection of antibodies for pre-clinical or clinical
testing on this basis. It
has been found that antibodies having an FAA amino acid motif in CDR3 of the
heavy chain are
particularly effective in cross-reacting with non-human primate CD3 proteins.
26

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
[00100] Engagement of the T cell receptor (TCR), either by binding MH-
peptide complexes or
anti-TCR/CD3 antibodies, initiates T cell activation. Examples of anti-TCR/CD3
antibodies that
activate T cells are OKT3 and UCHT1. These anti-CD3 antibodies cross-compete
for binding to
CD3 on T cells and are routinely used in T cell activation assays. Anti-CD3
antibodies of this
invention cross-compete with OKT3 for binding to human CD3. Depending on the
binding
affinity for CD3 and epitope on CD3, anti-CD3 antibodies activated T cells
with different
functional outcomes. In vitro incubation of human T cells with low affinity
anti-CD3 antibodies
resulted in incomplete activation of T cells, low IL-2 and IL-10 production.
In contrast, high-
affinity CD3 binders activated T cells to produce significantly more IL-2 and
other cytokines. The
low-affinity anti-CD3 antibodies are considered partial agonists that
selectively induce some
effector functions, potent tumor killing and 0D69 upregulation, while failing
to induce others,
such as IL-2 and IL-10 production. The high-affinity binders of this invention
are full-agonists
activating many immune effector functions of T cells. The strength of the
interaction with CD3
and the epitope recognized resulted in qualitatively different activation of T
cells. Maximal
cytokine production of T cells activated by low-affinity anti-CD3 antibodies
was lower than
maximal activation by high-affinity anti-CD3 antibodies. In some embodiments,
an antibody of
the invention results in a lower release of one or both of IL-2 and IL-10 when
combined with T
cells in an activation assay when compared to a reference anti-CD3 antibody in
the same
assay, where the reference antibody can be ID 304703 (or an antibody of
equivalent affinity.
The maximal release of IL-2 and/or IL-10 can be less than about 75% of the
release by the
reference antibody, less than about 50% of the release by the reference
antibody, less than
about 25% of the release by the reference antibody, and may be less than about
10% of the
release by a reference antibody.
[00101] In some embodiments of the invention, bispecific or multispecific
antibodies are
provided, which may have any of the configurations discussed herein, including
withotu
limitation a three chain bispecific. Bispecific antibodies comprise at least
the heavy chain
variable region of an antibody specific for a protein other than CD3, and may
comprise a heavy
and light chain variable region. In some such embodiments, the second antibody
specificity
binds to a tumor associated antigen, a targeting antigen, e.g. integrins,
etc., a pathogen antigen,
a checkpoint protein, and the like. Various formats of bispecific antibodies
are within the ambit
of the invention, including without limitation single chain polypeptides, two
chain polypeptides,
three chain polypeptides, four chain polypeptides, and multiples thereof.
[00102] The family of CD3 specific antibodies of the invention comprise a
VH domain,
comprising CDR1, CDR2 and CDR3 sequences in a human VH framework. The CDR
27

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
sequences may be situated, as an example, in the region of around amino acid
residues 26-35;
53-59; and 98-117 for CDR1, CDR2 and CDR3, respectively, of the provided
exemplary variable
region sequences set forth in SEQ ID NO:1-68. It will be understood by one of
skill in the art
that the CDR sequences may be in different position if a different framework
sequence is
selected, although generally the order of the sequences will remain the same.
[00103] The CDR sequences for an antibody of the invention may have the
following sequence
formulas. An X indicates a variable amino acid, which may be specific amino
acids as indicated
below.
CDR1
G1 G2 S3 14 X5 S6 X7 X8 X9 X10
where:
X5 may be any amino acid; in some embodiments X5 is S or R;
X7 and X8 may be any amino acid; in some embodiments X7 and X8 are,
independently,
S or G. In some embodiments X7X8 are SS, or GG;
X9 may be any amino acid; in some embodiments X9 is H or Y; in some
embodiments X9
is H;
X10 may be any amino acid, in some embodiments X10 is Y or F; in some
embodiments
X10 is Y.
In some embodiments, the CDR1 sequence has the formula: GG s I x5 SHFIG Y,
where X5 is as defined above. In some embodiments a CDR1 sequence of an anti-
CD3
antibody of the invention comprises the sequence set forth in any of SEQ ID
NO:1-68, residues
26-35.
CDR2
II' X2' X3, S4, Gs, X6, X7,
where:
X2, may be any amino acid; in some embodiments X2, is S, Y or H;
X3, may be any amino acid; in some embodiments X3, is Y, H or R;
X6, may be any amino acid; in some embodiments X6, is S, N or I or R;
X7, may be any amino acid; in some embodiments X7, is T or P.
In some embodiments the CDR2 sequence has the formula: I x2, x3, S G S T; or
I X2, x3, s GNP where X2, and X3, are as defined above. In some embodiments a
CDR2
sequence of an anti-CD3 antibody of the invention comprises the sequence set
forth in any of
SEQ ID NO:1-68, residues 53-59.
28

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
CDR3
X1" R2" W.3" R4" Hs" D6" 17" X8" X9" X10" Y11" P12" Y13" Y14" Y15" Y16" G17"
M18" D19" V20"
where:
X1,, may be any amino acid; in some embodiments X1,, is A or G.
X8,, may be any amino acid; in some embodiments X8,, is L or F.
X9,, may be any amino acid; in some embodiments X9,, is T or A
Xvy, may be any amino acid; in some embodiments Xvy, is G, A or R.
In some embodiments X8,, X9,, Xvy, are F A A, which motif corresponds to
antibodies that
cross-react with the CD3 protein of Cynomolgus macaque. In other embodiments
X8,, X9,, Xvy, are
L T A. In some embodiments a CDR3 sequence of an anti-CD3 antibody of the
invention
comprises the sequence set forth in any of SEQ ID NO:1-68, residues 98-117.
[00104] In some embodiments the CD3-binding VH domain is paired with a
light chain variable
region domain. In some such embodiments the light chain is a fixed light
chain. In some
embodiments the light chain comprises a VL domain with CDR1, CDR2 and CDR3
sequences
in a human VL framework. The CDR sequences may be those of SEQ ID NO:69. In
some
embodiments, the CDR1 sequence comprises amino acid residues 27-32; 50-52; 89-
97 for
CDR1, CDR2, CDR3, respectively.
[00105] In some embodiments the CDR sequences of an antibody of the
invention are a
sequence with at least 85% identity, at least 90% identity, at least 95%
identity, at least 99%
identity relative to a CDR sequence or set of CDR sequences in any one of SEQ
ID NO:1-68. In
some embodiments a CDR sequence of the invention comprises one, two, three or
more amino
acid substitutions relative to a CDR sequence or set of CDR sequences in any
one of SEQ ID
NO:1-68. In some embodiments said amino acid substitution(s) are one or more
of position 5 or
of CDR1, position 2, 6 or 7 of CDR2, position 1, 8, 9 or 10 of CDR3, relative
to the formulas
provided above.
[00106] Where a protein of the invention is a bispecific antibody, one
binding moiety, i.e. VH/VL
combination or VH only, is specific for human CD3 while the other arm may be
specific for target
cells, including cancer cells, such as cells of ovarian, breast,
gastrointestinal, brain, head and
neck, prostate, colon, and lung cancers, and the like, as well as hematologic
tumors such as B-
cell tumors, including leukemias, lymphomas, sarcomas, carcinomas, neural cell
tumors,
squamous cell carcinomas, germ cell tumors, metastases, undifferentiated
tumors, seminomas,
melanomas, myelomas, neuroblastomas, mixed cell tumors, neoplasias caused by
infectious
agents, and other malignancies, cells infected with a pathogen, autoreactive
cells causing
29

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
inflammation and/or autoimmunity. The non-CD3 moiety can also be specific for
an immune
regulatory protein, as will be described herein.
[00107] Tumor-associated antigens (TAAs) are relatively restricted to tumor
cells, whereas
tumor-specific antigens (TSAs) are unique to tumor cells. TSAs and TAAs
typically are portions
of intracellular molecules expressed on the cell surface as part of the major
histocompatibility
complex.
[00108] Tissue specific differentiation antigens are molecules present on
tumor cells and their
normal cell counterparts. Tumor-associated antigens known to be recognized by
therapeutic
mAbs fall into several different categories. Hematopoietic differentiation
antigens are
glycoproteins that are usually associated with cluster of differentiation (CD)
groupings and
include CD20, CD30, 0D33 and 0D52. Cell surface differentiation antigens are a
diverse group
of glycoproteins and carbohydrates that are found on the surface of both
normal and tumor
cells. Antigens that are involved in growth and differentiation signaling are
often growth factors
and growth factor receptors. Growth factors that are targets for antibodies in
cancer patients
include CEA, epidermal growth factor receptor (EGFR; also known as ERBB1)'
ERBB2 (also
known as HER2), ERBB3, MET (also known as HGFR), insulin-like growth factor 1
receptor
(IGF1R), ephrin receptor A3 (EPHA3), tumor necrosis factor (TNF)-related
apoptosis-inducing
ligand receptor 1 (TRAILR1; also known as TNFRSF10A), TRAILR2 (also known as
TNFRSF10B) and receptor activator of nuclear factor-KB ligand (RANKL; also
known as
TNFSF11). Antigens involved in angiogenesis are usually proteins or growth
factors that support
the formation of new microvasculature, including vascular endothelial growth
factor (VEGF),
VEGF receptor (VEGFR), integrin aV133 and integrin a5131. Tumor stroma and the
extracellular
matrix are indispensable support structures for a tumor. Stromal and
extracellular matrix
antigens that are therapeutic targets include fibroblast activation protein
(FAP) and tenascin.
[00109] Examples of therapeutic antibodies useful in bispecific
configurations include, without
limitation, rituximab; lbritumomab; tiuxetan; tositumomab; Brentuximab;
vedotin; Gemtuzumab;
ozogamicin; Alemtuzumab; IGN101; adecatumumab; Labetuzumab; huA33; Pemtumomab;

oregovomab; CC49 (minretumomab); cG250; J591; MOv18; MORAb-003 (farletuzumab);
3F8,
ch14.18; KW-2871; hu3S193; IgN311; Bevacizumab; IM-2C6; CDP791; Etaracizumab;
Volociximab; Cetuximab, panitumumab, nimotuzumab; 806; Trastuzumab;
pertuzumab; MM-
121; AMG 102, METMAB; SCH 900105; AVE1642, IMC-Al2, MK-0646, R1507; CP 751871;

KB004; II 1A4; Mapatumumab (HGS-ETR1); HGS-ETR2; CS-1008; Denosumab;
Sibrotuzumab;
F19; and 8106.

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
[00110] The immune-checkpoint receptors that have been most actively
studied in the context of
clinical cancer immunotherapy, cytotoxic T-lymphocyte-associated antigen 4
(CTLA4; also
known as 0D152) and programmed cell death protein 1 (PD1; also known as 0D279)
- are both
inhibitory receptors. The clinical activity of antibodies that block either of
these receptors implies
that antitumor immunity can be enhanced at multiple levels and that
combinatorial strategies
can be intelligently designed, guided by mechanistic considerations and
preclinical models.
[00111] The two ligands for PD1 are PD1 ligand 1 (PDL1; also known as B7-H1
and 0D274) and
PDL2 (also known as B7-DC and 0D273). PDL1 is expressed on cancer cells and
through
binding to its receptor PD1 on T cells it inhibits T cell activation/function.
[00112] Lymphocyte activation gene 3 (LAG3; also known as 0D223), 2B4 (also
known as
0D244), B and T lymphocyte attenuator (BTLA; also known as 0D272), T cell
membrane
protein 3 (TIM3; also known as HAVcr2), adenosine A2a receptor (A2aR) and the
family of killer
inhibitory receptors have each been associated with the inhibition of
lymphocyte activity and in
some cases the induction of lymphocyte anergy. Antibody targeting of these
receptors can be
used in the methods of the invention.
[00113] Agents that agonize an immune costimulatory molecule are also
useful in the methods of
the invention. Such agents include agonists or CD40 and 0X40. CD40 is a
costimulatory
protein found on antigen presenting cells (APCs) and is required for their
activation. These
APCs include phagocytes (macrophages and dendritic cells) and B cells. CD40 is
part of the
TNF receptor family. The primary activating signaling molecules for CD40 are
IFNy and CD40
ligand (CD4OL). Stimulation through CD40 activates macrophages.
[00114] Anti CCR4 (0D194) antibodies of interest include humanized
monoclonal antibodies
directed against C-C chemokine receptor 4 (CCR4) with potential anti-
inflammatory and
antineoplastic activities. CCR2 is expressed on inflammatory macrophages that
can be found in
various inflammatory conditions, e.g. rheumatoid arthritis; and have also been
identified as
expressed on tumor promoting macrophages. CCR2 is also expressed on regulatory
T cells,
and the CCR2 ligand, CCL2, mediates recruitment of regulatory T cells into
tumors. Regulatory
T cells suppress a response for anti-tumor T cells and thus their inhibition
or depletion is
desired.
PRODUCING PROTEINS OF THE INVENTION
[00115] Although antibodies can be prepared by chemical synthesis, they are
typically produced
by methods of recombinant DNA technology, such as co-expression of all the
chains making up
31

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
the protein in a single recombinant host cell, or co-expression of a heavy
chain polypeptide and
an antibody, e.g. a human antibody. In addition, the antibody heavy and light
chains can also be
expressed using a single polycistronic expression vector. Purification of
individual polypeptides
is achieved using standard protein purification technologies such as affinity
(protein A)
chromatography, size exclusion chromatography and/or hydrophobic interaction
chromatography. Bispecifics are sufficiently different in size and
hydrophobicity that purification
can be performed using standard procedures.
[00116] The amount of antibody and heavy chain polypeptide produced in a
single host cell can
be minimized through engineering of constant regions of the antibody and the
heavy chain such
that homodimerization is favored over heterodimerization, e.g. by introducing
self-
complementary interactions (see e.g. WO 98/50431 for possibilities, such as
"protuberance-into-
cavity" strategies (see WO 96/27011)). It is therefore another aspect of the
present invention to
provide a method for producing a bispecific in a recombinant host, the method
including the step
of: expressing in a recombinant host cell a nucleic acid sequences encoding at
least two heavy
chain polypeptides, wherein said heavy chain polypeptides differ in their
constant regions
sufficiently to reduce or prevent homodimer formation but increase bispecific
formation.
[00117] Where the protein comprises three chains, e.g. FlicAbs, they may be
produced by co-
expression of the three chains (2 heavy chains and one light chain) making up
the molecule in a
single recombinant host cell.
[00118] For recombinant production of the proteins herein, one or more
nucleic acids encoding
all chains, e.g. 2, 3 4, etc. are isolated and inserted into a replicable
vector for further cloning
(amplification of the DNA) or for expression. Many vectors are available. The
vector
components generally include, but are not limited to, one or more of the
following: a signal
sequence, an origin of replication, one or more marker genes, an enhancer
element, a
promoter, and a transcription termination sequence.
[00119] In a preferred embodiment, the host cell according to the method of
the invention is
capable of high-level expression of human immunoglobulin, i.e. at least 1
pg/cell/day, preferably
at least 10 pg/cell/day and even more preferably at least 20 pg/cell/day or
more without the
need for amplification of the nucleic acid molecules encoding the single
chains in said host cell.
PHARMACEUTICAL COMPOSITION
[00120] It is another aspect of the present invention to provide
pharmaceutical compositions
comprising one or more proteins of the present invention in admixture with a
suitable
pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers as
used herein are
32

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
exemplified, but not limited to, adjuvants, solid carriers, water, buffers, or
other carriers used in
the art to hold therapeutic components, or combinations thereof.
[00121] Therapeutic formulations of the proteins used in accordance with
the present invention
are prepared for storage by mixing proteins having the desired degree of
purity with optional
pharmaceutically acceptable carriers, excipients or stabilizers (see, e.g.
Remington's
Pharmaceutical Sciences 16th edition, Osol, A. Ed. (1980)), such as in the
form of lyophilized
formulations or aqueous solutions. Acceptable carriers, excipients, or
stabilizers are nontoxic to
recipients at the dosages and concentrations employed, and include buffers
such as phosphate,
citrate, and other organic acids; antioxidants including ascorbic acid and
methionine;
preservatives (such as octadecyldimethylbenzyl ammonium chloride;
hexamethonium chloride;
benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol;
alkyl parabens
such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-
pentanol; and m-cresol);
low molecular weight (less than about 10 residues) polypeptides; proteins,
such as serum
albumin, gelatin, or immunoglobulins; hydrophilic polymers such as
polyvinylpyrrolidone; amino
acids such as glycine, glutamine, asparagine, histidine, arginine, or lysine;
monosaccharides,
disaccharides, and other carbohydrates including glucose, mannose, or
dextrins; chelating
agents such as EDTA; sugars such as sucrose, mannitol, trehalose or sorbitol;
salt-forming
counter-ions such as sodium; metal complexes (e.g. Zn-protein complexes);
and/or non-ionic
surfactants such as TWEEN TM, PLURONICSTM or polyethylene glycol (PEG).
[00122] Anti-CD3 antibody formulations are disclosed, for example, in U.S.
Patent Publication
No. 20070065437, the entire disclosure is expressly incorporated by reference
herein. Similar
formulations can be used for the proteins of the present invention. The main
components of
such formulations are a pH buffering agent effective in the range of 3.0 to
6.2, a salt, a
surfactant, and an effective amount of a bispecific with anti-CD3 specificity.
METHODS OF USE
[00123] Methods are provided for treating or reducing disease, including
without limitation
infection, autoimmune disease, primary or metastatic cancer, etc. in a regimen
comprising
contacting the targeted cells with an antigen-binding composition of the
invention, particularly
where the antigen-binding composition is a multi-specific antibody suitable
for the condition
being treated, e.g. where one binding moiety specifically binds to a tumor
associated antigen for
treatment of the relevant cancer cells; a binding moiety specific for a
pathogen of interest for
treatment of the relevant infection, and the like. Such methods include
administering to a
subject in need of treatment a therapeutically effective amount or an
effective dose of the
33

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
agents of the invention, including without limitation combinations of the
reagent with a
chemotherapeutic drug, radiation therapy, or surgery.
[00124]
Effective doses of the compositions of the present invention for the
treatment of disease
vary depending upon many different factors, including means of administration,
target site,
physiological state of the patient, whether the patient is human or an animal,
other medications
administered, and whether treatment is prophylactic or therapeutic. Usually,
the patient is a
human, but nonhuman mammals may also be treated, e.g. companion animals such
as dogs,
cats, horses, etc., laboratory mammals such as rabbits, mice, rats, etc., and
the like. Treatment
dosages can be titrated to optimize safety and efficacy.
[00125]
Dosage levels can be readily determined by the ordinarily skilled clinician,
and can be
modified as required, e.g., as required to modify a subject's response to
therapy. The amount of
active ingredient that can be combined with the carrier materials to produce a
single dosage
form varies depending upon the host treated and the particular mode of
administration. Dosage
unit forms generally contain between from about 1 mg to about 500 mg of an
active ingredient.
[00126]
In some embodiments, the therapeutic dosage the agent may range from about
0.0001
to 100 mg/kg, and more usually 0.01 to 5 mg/kg, of the host body weight. For
example dosages
can be 1 mg/kg body weight or 10 mg/kg body weight or within the range of 1-10
mg/kg. An
exemplary treatment regime entails administration once every two weeks or once
a month or
once every 3 to 6 months.
Therapeutic entities of the present invention are usually
administered on multiple occasions. Intervals between single dosages can be
weekly, monthly
or yearly. Intervals can also be irregular as indicated by measuring blood
levels of the
therapeutic entity in the patient. Alternatively, therapeutic entities of the
present invention can
be administered as a sustained release formulation, in which case less
frequent administration
is required. Dosage and frequency vary depending on the half-life of the
polypeptide in the
patient.
[00127]
In prophylactic applications, a relatively low dosage may be administered at
relatively
infrequent intervals over a long period of time. Some patients continue to
receive treatment for
the rest of their lives. In other therapeutic applications, a relatively high
dosage at relatively
short intervals is sometimes required until progression of the disease is
reduced or terminated,
and preferably until the patient shows partial or complete amelioration of
symptoms of disease.
Thereafter, the patent can be administered a prophylactic regime.
[00128]
In still other embodiments, methods of the present invention include
treating, reducing or
preventing tumor growth, tumor metastasis or tumor invasion of cancers
including carcinomas,
hematologic cancers such as leukemias and lymphomas, melanomas, sarcomas,
gliomas, etc.
34

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
For prophylactic applications, pharmaceutical compositions or medicaments are
administered to
a patient susceptible to, or otherwise at risk of disease in an amount
sufficient to eliminate or
reduce the risk, lessen the severity, or delay the outset of the disease,
including biochemical,
histologic and/or behavioral symptoms of the disease, its complications and
intermediate
pathological phenotypes presenting during development of the disease.
[00129]
Compositions for the treatment of disease can be administered by parenteral,
topical,
intravenous, intratumoral, oral, subcutaneous, intraarterial, intracranial,
intraperitoneal,
intranasal or intramuscular means.
A typical route of administration is intravenous or
intratumoral, although other routes can be equally effective.
[00130]
Typically, compositions are prepared as injectables, either as liquid
solutions or
suspensions; solid forms suitable for solution in, or suspension in, liquid
vehicles prior to
injection can also be prepared. The preparation also can be emulsified or
encapsulated in
liposomes or micro particles such as polylactide, polyglycolide, or copolymer
for enhanced
adjuvant effect, as discussed above. Langer, Science 249: 1527, 1990 and
Hanes, Advanced
Drug Delivery Reviews 28: 97-119, 1997. The agents of this invention can be
administered in
the form of a depot injection or implant preparation which can be formulated
in such a manner
as to permit a sustained or pulsatile release of the active ingredient. The
pharmaceutical
compositions are generally formulated as sterile, substantially isotonic and
in full compliance
with all Good Manufacturing Practice (GMP) regulations of the U.S. Food and
Drug
Administration.
[00131]
Toxicity of the proteins described herein can be determined by standard
pharmaceutical
procedures in cell cultures or experimental animals, e.g., by determining the
LD50 (the dose
lethal to 50% of the population) or the LD100 (the dose lethal to 100% of the
population). The
dose ratio between toxic and therapeutic effect is the therapeutic index. The
data obtained from
these cell culture assays and animal studies can be used in formulating a
dosage range that is
not toxic for use in human. The dosage of the proteins described herein lies
preferably within a
range of circulating concentrations that include the effective dose with
little or no toxicity. The
dosage can vary within this range depending upon the dosage form employed and
the route of
administration utilized. The exact formulation, route of administration and
dosage can be chosen
by the individual physician in view of the patient's condition.
[00132]
The pharmaceutical compositions can be administered in a variety of unit
dosage forms
depending upon the method of administration. For example, unit dosage forms
suitable for oral
administration include, but are not limited to, powder, tablets, pills,
capsules and lozenges. It is
recognized that compositions of the invention when administered orally, should
be protected

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
from digestion. This is typically accomplished either by complexing the
molecules with a
composition to render them resistant to acidic and enzymatic hydrolysis, or by
packaging the
molecules in an appropriately resistant carrier, such as a liposome or a
protection barrier.
Means of protecting agents from digestion are well known in the art.
[00133] The compositions for administration will commonly comprise an
antibody or other
ablative agent dissolved in a pharmaceutically acceptable carrier, preferably
an aqueous carrier.
A variety of aqueous carriers can be used, e.g., buffered saline and the like.
These solutions are
sterile and generally free of undesirable matter. These compositions may be
sterilized by
conventional, well known sterilization techniques. The compositions may
contain
pharmaceutically acceptable auxiliary substances as required to approximate
physiological
conditions such as pH adjusting and buffering agents, toxicity adjusting
agents and the like, e.g.,
sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium
lactate and the
like. The concentration of active agent in these formulations can vary widely,
and will be
selected primarily based on fluid volumes, viscosities, body weight and the
like in accordance
with the particular mode of administration selected and the patient's needs
(e.g., Remington's
Pharmaceutical Science (15th ed., 1980) and Goodman & Gillman, The
Pharmacological Basis
of Therapeutics (Hardman et al., eds., 1996)).
[00134] Also within the scope of the invention are kits comprising the
active agents and
formulations thereof, of the invention and instructions for use. The kit can
further contain a least
one additional reagent, e.g. a chemotherapeutic drug, etc. Kits typically
include a label
indicating the intended use of the contents of the kit. The term label
includes any writing, or
recorded material supplied on or with the kit, or which otherwise accompanies
the kit.
[00135] The compositions can be administered for therapeutic treatment.
Compositions are
administered to a patient in an amount sufficient to substantially ablate
targeted cells, as
described above. An amount adequate to accomplish this is defined as a
"therapeutically
effective dose.", which may provide for an improvement in overall survival
rates. Single or
multiple administrations of the compositions may be administered depending on
the dosage and
frequency as required and tolerated by the patient. The particular dose
required for a treatment
will depend upon the medical condition and history of the mammal, as well as
other factors such
as age, weight, gender, administration route, efficiency, etc.
[00136] The invention now being fully described, it will be apparent to one
of ordinary skill in the
art that various changes and modifications can be made without departing from
the spirit or
scope of the invention.
36

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
EXAMPLES
Example 1
Genetically Engineered Rats Expressing Heavy Chain-Only Antibodies
[00137] A human IgH locus was constructed and assembled in several parts,
which involved the
modification and joining of rat C region genes, which were then joined
downstream of human
VH6-D-JH region. Two BACs with separate clusters of human VH genes were then
co-injected
with a BAC encoding the assembled (human VH6-D-JH-rat C) fragment.
[00138] Transgenic rats carrying artificial heavy chain immunoglobulin loci
in unrearranged
configuration were generated. The included constant region genes encode IgM,
IgD, IgG2b,
IgE, IgA and 3' enhancer. RT-PCR and serum analysis (ELISA) of transgenic rats
revealed
productive rearrangement of transgenic immunoglobulin loci and expression of
heavy chain only
antibodies of various isotypes in serum. Transgenic rats were cross-bred with
rats with mutated
endogenous heavy chain and light chain loci previously described in US patent
publication
2009/0098134 Al. Analysis of such animals demonstrated inactivation of rat
immunoglobulin
heavy and light chain expression and high level expression of heavy chain
antibodies with
variable regions encoded by human V, D, and J genes. Immunization of
transgenic rats resulted
in production of high titer serum responses of antigen-specific heavy chain
antibodies. These
transgenic rats expressing heavy chain antibodies with a human VDJ region were
called
UniRats.
Example 2
Genetically Engineered Rats Expressing Fixed Light Chain Antibodies
[00139] Transgenic human antibody repertoires were generated from H-chains
with diverse (VH-
D-JH)n rearrangement in combination with a unique L-chain. For this a
rearranged L-chain,
human Vk-Jkl-Ck, was integrated in the rat germline by DNA microinjection and
the obtained
transgenic animals were bred with a previously described rat strain that
expresses a human H-
chain repertoire naturally (Osborn etal., 2013). This new rat strain was named
OmniFlic.
[00140] Immunizations of OmniFlic rats, using many different antigens,
produced high levels of
antigen-specific IgG similar to other transgenic rats carrying the same IgH
locus. Repertoire
analysis by RT-PCR identified highly variable VH-gene rearrangements at high
transcript and
protein levels. In addition, only one L-chain product, also expressed at high
level, was identified.
[00141] Antigen-specific binders from OmniFlic were obtained by NGS and
selection from cDNA
libraries (yeast, E.coli, phage), which upon sequencing identified diverse H-
chain transcripts.
37

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
For the expression in mammalian cells hypermutated H-chain constructs were
transfected in
combination with the original transgenic lgic sequence. In this rearranged Vk-
Jk1-Ck no
mutational changes were allowed and always the same L-chain was expressed with
various H-
chain products to generate monoclonal human IgG.
Example 3
Generation of Antigen-Specific Antibodies in Transgenic Rats,
[00142] For the generation of antigen-specific heavy chain antibodies in
rats, genetically
engineered rats expressing were immunized in two ways.
[00143] Immunization with recombinant extracellular domains of PD-L1 and BCMA.

Recombinant extracellular domains of PD-L1 and BCMA were purchased from R&D
Systems
and were diluted with sterile saline and combined with adjuvant. lmmunogens
were either
combined with Complete Freund's Adjuvant (CFA) and Incomplete Freund's
Adjuvant (IFA) or
Titermax and Ribi adjuvants. The first immunization (priming) with immunogen
in CFA or
Titermax was administered in the left and right legs. After the first
immunization with
immunogens in CFA two more immunizations in IFA (boosters) or 4 more
immunizations in Ribi
and one more in Titermax were administered in each leg. This sequence of
immunizations leads
to the development of B cells producing high affinity antibodies. The
immunogen concentrations
were 10 microgram per leg. Serum was collected from rats at the final bleed to
determine serum
titers.
[00144] For the generation of anti-human CD386 antibodies genetically
engineered rats were
immunized using DNA-based immunization protocols.
[00145] OmniFlic rats were immunized with human and cynomolgus CD3-
epsilon/delta
constructs at Aldevron, Inc. (Fargo, ND) using the GENOVAC Antibody
Technology. Draining
lymph nodes were harvested after the final boost and RNA isolated. Following
cDNA synthesis,
the IgH heavy chain antibody repertoire was characterized by Next Generation
Sequencing and
our proprietary in-house software. Candidate antigen-specific VH sequences
showing evidence
of antigen-specific positive selection were selected. Several hundred VH
sequences encoding
FlicAbs were selected for gene assembly and cloned into an expression vector.
Subsequently,
fully human FlicAb IgG1 antibodies were expressed in HEK cells for analysis by
Flow and
ELISA. Human FlicAbs were tested for binding to primary human T cells and
Jurkat cells by
flow. In addition, human FlicAbs were tested using recombinant CD386 proteins
in ELISA. All
FlicAbs with positive binding for human T cells are listed in Figure 1.
Selected sequences were
further characterized in T cell activation assays.
38

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
Example 4 Characterization of anti-CD3 OmniFlic Antibodies
[00146]
Antibodies derived from the campaign as described above were further
characterized for
their ability to activate T cells to produce cytokines and to upregulate CD69
on its surface. For
the quantitation of the IL-2 produced by Jurkat cells cells or peripheral
blood lymphocytes
BioLegend's ELISA max kit was used. Results of the experiment using human
peripheral blood
T cells and different concentrations of OmniFlic antibodies is shown in Figure
3.
Example 5 Characterization of Bispecific Antibodies
[00147]
The anti-CD3 FlicAb ID 304703 (SEQ ID NO:39) was selected for further
development in
bispecifics. This FlicAb cross-reacts with cynomolgus CD3 and stimulates human
T cells
potently. Two types of bispecifics were produced (see Figure 2 for schematic
representation).
Knobs-into-holes technology was used to generate bispecific FlicAbs (Protein
Engineering vol.9
no.7 pp.617-621, 1996, 'Knobs-into-holes' engineering of antibody CH3 domains
for heavy
chain heterodimerization. John B.B.Ridgway, Leonard G.Presta and Paul Carter).
The C-
terminus of the heavy chain with the knob was tagged with a C-tag and
heterodimeric antibodies
were purified using the CaptureSelect C-Tag Affinity Matrix (Thermo Fischer
Scientific). A
bispecific FlicAb with one arm reacting with CD3 (ID 304703) and the other
with human PD-L1
was produced and was shown to activate human CD8+ T cells only in the presence
of PD-L1
positive tumor cells (Figure 3). HDLM2 is a Multiple Myeloma cell line, which
expresses PD-L1
on the surface. Ramos is a Burkitt's lymphoma cell line, which is negative for
PD-L1. CD69
expression was used as a read-out.
Bispecific antibody was used at the indicated
concentrations.
[00148]
As shown in Figure 4, tumor cells (HDLM2, which express PD-L1 on the cell
surface)
were incubated with purified human CD8+ T cells and bispecific antibodies.
HDLM2 cells do not
express CD20 and co-culture with an a-CD3/a-CD20 bispecific FlicAb did not
lead to killing of
HDLM2 cells. Only co-culture of human CD8+ T cells and HDLM2 with an a-CD3/a-
PD-L1
bispecific FlicAb led to significant killing.
[00149]
Figure 5 summarizes data for antibodies in monospecific and bispecific
format. Column
1 shows the sequence ID for the anti-CD3 VH sequence ID NO:304703 (SEQ ID
NO:39),
314171 (SEQ ID NO:13), 313306 (SEQ ID NO:1), 313329 (SEQ ID NO:6) and 313283
(SEQ ID
NO:18). Column 2 shows the MFI value for Jurkat cell binding of the parental
monospecific anti-
CD3. Column 3 shows the MFI value for cyno T-cell binding of the parental
monospecific anti-
CD3. Column 5 shows the picograms of IL-2 released by pan T-cells stimulated
by the bispecific
39

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
antibody binding the BCMA protein coated on plastic at the dose indicated.
Column 6 shows
the picograms of IL-6 released by pan T-cells stimulated by the bispecific
antibody binding the
BCMA protein coated on plastic at the dose indicated. Column 7 shows the
picograms of IL-10
released by pan T-cells stimulated by the bispecific antibody binding the BCMA
protein coated
on plastic at the dose indicated. Column 8 shows the picograms of IFN-y
released by pan T-
cells stimulated by the bispecific antibody binding the BCMA protein coated on
plastic at the
dose indicated. Column 9 shows the picograms of TNFa released by pan T-cells
stimulated by
the bispecific antibody binding the BCMA protein coated on plastic at the dose
indicated.
Column 10 shows the EC50 of bispecific antibody-mediated U266 tumor cell lysis
in presence of
human pan T-cells. Column 11 shows the percent lysis of U266 tumor cells in
the presence of
bispecific antibody and human pan T-cells at a dose of 333ng/mL of bispecific
antibody.
Column 12 shows the protein binding affinity of the anti-CD3 arm of the
bispecific antibody
measured by Octet. Column 13 shows the MFI value for Jurkat cell binding of
the bispecific
antibody.
Example 6
Correlation of Bispecific Killing Activity and Cytokine release
[00150] Shown in Figure 6, four aCD3_famtaBCMA bispecific antibodies, each
with a unique
anti-CD3 arm (as indicated) and a common anti-BCMA arm, were tested for the
ability to kill
U266 BCMA+ tumor cells through redirection of activated primary T cells. In
this experiment
U266 cells that express BCMA were mixed with activated pan T-cells in a 10:1
E:T ratio along
with the addition of bispecific antibody. The x-axis shows the concentration
of antibody used
and the y-axis shows the % lysis of tumor cells 6 hours after addition of
antibody. The killing
activity was correlated with IL-2 release (Figure 7); with IFN-y release
(Figure 8) and with CD3
binding affinity (Figure 9). The correlation between IL-2 production and U266
tumor cell lysis is
R2= 0.37. The correlation between IFN-y production and U266 tumor cell lysis
is R2= 0.53. The
correlation between U266 killing EC50 and protein binding affinity is R2=
0.93.
[00151] aCD3_F1F:aBCMA bispecific antibodies were assayed for the ability
to kill three
different BCMA+ tumor cells and one BCMA-negative cell line through
redirection of activated
primary T cells. The antibodies were comprised of an aCD3 arm (SEQ ID NO:1 and
SEQ ID
NO:69) and an aBCMA arm (SEQ ID NO:70, or SEQ ID NO:71) (shown in Figures 10-
12). In
this experiment, tumor cells were mixed with activated pan T-cells in a 10:1
E:T ratio along with
the addition of bispecific antibody. Figure 10A shows killing of RPMI-8226
cells, Figure 10B

CA 03029209 2018-12-21
WO 2017/223111
PCT/US2017/038373
shows killing of NCI-H929 cells, panel C shows killing of U-266 cells, and
Figure 10D shows
killing of K562 cells, a negative control. The x-axis shows the concentration
of antibody used
and the y-axis shows the % lysis of tumor cells 6 hours after addition of
antibody.
[00152] Figure 11 shows the level of IL-2 cytokine release was measured
after resting human T
cells were cultured with various tumor cell lines and increasing doses of
aCD3_F1F:aBCMA
bispecific antibody (as in Figure 10). Figure 11A shows IL-2 release
stimulated by RPMI-8226
cells, Figure 11B shows IL-2 release stimulated by NCI-H929 cells, Figure 110
shows IL-2
release stimulated by U-266 cells, and Figure 11D shows IL-2 release
stimulated by K562 cells,
a negative control.
[00153] The level of IFN-y cytokine release was measured after resting
human T cells were
cultured with various tumor cell lines and increasing doses of aCD3_F1F:aBCMA
bispecific
antibody (as in Figure 10). Figure 12A shows IFN-y release stimulated by RPMI-
8226 cells,
Figure 12B shows IFN-y release stimulated by NCI-H929 cells, Figure 120 shows
IFN-y release
stimulated by U-266 cells, and Figure 12D shows IFN-y release stimulated by
K562 cells, a
negative control.
[00154] The examples are put forth so as to provide those of ordinary skill
in the art with a
complete disclosure and description of how to make and use the present
invention, and are not
intended to limit the scope of what the inventors regard as their invention
nor are they intended
to represent that the experiments below are all or the only experiments
performed. Efforts have
been made to ensure accuracy with respect to numbers used (e.g. amounts,
temperature, etc.)
but some experimental errors and deviations should be accounted for. Unless
indicated
otherwise, parts are parts by weight, molecular weight is weight average
molecular weight,
temperature is in degrees Centigrade, and pressure is at or near atmospheric.
[00155] While the present invention has been described with reference to
the specific
embodiments thereof, it should be understood by those skilled in the art that
various changes
may be made and equivalents may be substituted without departing from the true
spirit and
scope of the invention. In addition, many modifications may be made to adapt a
particular
situation, material, composition of matter, process, process step or steps, to
the objective, spirit
and scope of the present invention. All such modifications are intended to be
within the scope
of the claims appended hereto.
41

Representative Drawing
A single figure which represents the drawing illustrating the invention.
Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date Unavailable
(86) PCT Filing Date 2017-06-20
(87) PCT Publication Date 2017-12-28
(85) National Entry 2018-12-21
Examination Requested 2022-06-20

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $277.00 was received on 2024-05-21


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if standard fee 2025-06-20 $277.00
Next Payment if small entity fee 2025-06-20 $100.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Registration of a document - section 124 $100.00 2018-12-21
Application Fee $400.00 2018-12-21
Maintenance Fee - Application - New Act 2 2019-06-20 $100.00 2019-05-22
Maintenance Fee - Application - New Act 3 2020-06-22 $100.00 2020-05-26
Maintenance Fee - Application - New Act 4 2021-06-21 $100.00 2021-05-28
Maintenance Fee - Application - New Act 5 2022-06-20 $203.59 2022-05-25
Request for Examination 2022-06-20 $814.37 2022-06-20
Maintenance Fee - Application - New Act 6 2023-06-20 $210.51 2023-05-24
Maintenance Fee - Application - New Act 7 2024-06-20 $277.00 2024-05-21
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
TENEOBIO, INC.
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Request for Examination / Amendment 2022-06-20 11 329
Claims 2018-12-21 5 226
Claims 2022-06-20 5 208
Office Letter 2023-04-14 1 192
Abstract 2018-12-21 1 63
Claims 2018-12-21 5 159
Drawings 2018-12-21 11 333
Description 2018-12-21 41 2,380
Representative Drawing 2018-12-21 1 4
Patent Cooperation Treaty (PCT) 2018-12-21 2 83
International Search Report 2018-12-21 5 341
Declaration 2018-12-21 2 106
National Entry Request 2018-12-21 8 264
Voluntary Amendment 2018-12-21 7 203
Cover Page 2019-01-10 1 32
Examiner Requisition 2023-06-22 5 284
Amendment 2023-10-20 32 1,422
Description 2023-10-20 41 3,386
Claims 2023-10-20 5 279
Drawings 2023-10-20 12 740

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :